CN113354590A - Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway - Google Patents

Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway Download PDF

Info

Publication number
CN113354590A
CN113354590A CN202010146043.6A CN202010146043A CN113354590A CN 113354590 A CN113354590 A CN 113354590A CN 202010146043 A CN202010146043 A CN 202010146043A CN 113354590 A CN113354590 A CN 113354590A
Authority
CN
China
Prior art keywords
compound
oxo
phenoxy
trifluoromethyl
dimethylamino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010146043.6A
Other languages
Chinese (zh)
Inventor
刘刚
马瑶
杨景舒
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ningbo Kangbai Ruige Medical Technology Co ltd
Original Assignee
Ningbo Kangbai Ruige Medical Technology Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ningbo Kangbai Ruige Medical Technology Co ltd filed Critical Ningbo Kangbai Ruige Medical Technology Co ltd
Priority to CN202010146043.6A priority Critical patent/CN113354590A/en
Publication of CN113354590A publication Critical patent/CN113354590A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/88Oxygen atoms
    • C07D239/90Oxygen atoms with acyclic radicals attached in position 2 or 3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/88Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Abstract

The invention discloses quinazolinone compounds antagonizing NOD1/2 receptor signal pathways, in particular quinazolinone compound micromolecule compounds shown in general formulas I and II, a preparation method of the quinazolinone compounds, a pharmaceutical composition containing the compounds, and activation of the compounds through antagonizing NOD1/2 signal transduction pathways, can be used for immunotherapy of organisms, especially for tumor therapy, and belongs to the technical field of medicines.

Description

Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway
Technical Field
The invention relates to novel quinazolinone small molecular compounds, a preparation method of the quinazolinone small molecular compounds, a pharmaceutical composition containing the compounds, and activation of the compounds through antagonism NOD1/2 signal transduction pathway, can be used for immunotherapy of organisms, especially for tumor therapy, and belongs to the technical field of medicines.
Background
Muramyl Dipeptide (MDP), a minimal glycopeptide fragment with immunoadjuvant activity (fig. 1, compound 1), was first found in the cell wall of mycobacteria, and can effectively increase the humoral and cellular immune responses of the body to antigens by NOD2 that non-specifically activates human immune cells (e.g., macrophages, myelomonocytes, neutrophils, T and B lymphocytes, etc.), thereby exerting non-specific anti-infection (e.g., pneumococci, escherichia coli, pseudomonas aeruginosa, mononucleosis, candida albicans, etc.), anti-tumor (fibrosarcoma, hepatoma, etc.), immunomodulation, etc.
Pharmaceutical chemists have completed a great deal of structural optimization for MDP molecules and summarized the structure-activity relationship of the system, and several molecules have been used as immunopotentiators to carry out clinical research or to obtain approval for marketing. For example, Murabutide (FIG. 1, Compound 2) is a derivative of the C-terminal fatty alkane of MDP molecule, and has been studied clinically in anti-AIDS field because it can enhance the nonspecific antibacterial and antiviral activities of host immune system, induce the body to release CSF, and regulate macrophage function. MDP-Lys (L18) (trade name Romultide or Muroctasin, FIG. 1, Compound 3) is a derivative obtained by connecting 18-carbon alkane aliphatic hydrocarbon to the side chain of MDP molecule through lysine (Lys) as a connecting bridge (linker). One phase of clinical experimental research shows that the compound can effectively increase the level of various cytokines (such as CSF, IL-1, IL6, IL8, MCP-1 and TNF-alpha) in vivo. The compound is finally approved to be on the market in Japan and is used for treating leukopenia caused by radiotherapy and chemotherapy, and the main adverse reactions are allergy, fever and shivering, and then the compound is removed from the market due to toxic and side effects. Mifamurtide (fig. 1, compound 4) is a prodrug of MDP conjugated with long chain aliphatic hydrocarbons via phosphatidyl group using alanine as linker, which was approved for osteosarcoma adjuvant chemotherapy (mainly ifosfamide) in europe in 2005, with major side effects of fever and chills.
Recently, the applicant has found that MDP-C (FIG. 1, compound 5), an immunological adjuvant, is an MDP derivative obtained by linking an MDP molecule to an aromatic conjugate system by a solid phase synthesis method using Lys as a linker. Compared with MDP, MDP-C has the characteristics of low heat source, no sensitization, low toxicity and the like, and can be used for preparing HBV therapeutic vaccines. Because the MDP-C compound still contains sugar structural fragments, the synthesis route is complex, the period is long and the cost is high. Further research shows that active pharmacophore D-Gln (light color part in figure 2) in MDP molecules is retained, sugar-containing muramyl is replaced by o-nitrobenzoyl or its derivative, and L-alanyl is replaced by other liposoluble amino acid acyl (figure 2) to retain better immunoadjuvant activity. This finding not only greatly reduces the cost of synthesis of this class of compounds, but also suggests that the sugar structure is not a fragment essential for MDP activity. Based on the above, the inventors have aimed at designing and synthesizing small molecule heterocyclic compounds with "dominant structures", and have found that two classes of small molecule heterocyclic compounds can still agonize or antagonize NOD1/2 signal pathways, including 1, 4-diazepine compounds and quinazolinone compounds. FIG. 3 shows the structure of the newly discovered lead compound 4h of 1, 4-diazepine and its antagonism IC to NOD1/250The compound can be used for sensing paclitaxel and obviously inhibiting the growth of Lewis lung cancer of mice.
The integration of D-Gln into the 1, 4-benzodiazepine-2, 5-dione structure forms the D-Gln-BZD backbone compound (FIG. 3,4 h). Exciting, the compounds show remarkable sensitization of paclitaxel against tumors in animals. The quinazolinone backbone molecules reported in this patent also have the dual antagonistic activity of NOD 1/2.
Disclosure of Invention
The invention relates to a compound of general formula (I) and (II), a stereoisomer and a pharmaceutically acceptable salt thereof, which are used as medicaments.
The second aspect of the present invention relates to a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of the general formulae (I), (II), a stereoisomer, a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
The third aspect of the invention relates to a compound of general formula (I) and (II), a stereoisomer and a pharmaceutically acceptable salt thereof, and application of the compound in preparing a medicament for preventing or treating immunoinflammatory diseases (including tumors), in particular immunoinflammatory diseases acting on NOD1/2 signaling pathways.
Specifically, the invention discloses a compound with a structure shown in a formula (I) or (II), a stereoisomer and a pharmaceutically acceptable salt thereof,
Figure BDA0002400750460000021
Figure BDA0002400750460000031
wherein:
R1、R2independently selected from hydrogen, C1-6Alkyl radical, R1、R2Can form a quaternary, five-membered or six-membered aliphatic heterocyclic ring with nitrogen at position 1 and/or carbon atom at position 2 of the mother nucleus of the compound of the general formula II independently or together; the quaternary, quinary or hexahydric aliphatic heterocyclic ring can be substituted or unsubstituted, and the substituent on the quaternary, quinary or hexahydric aliphatic heterocyclic ring can be one or more and is independently selected from the following groups: o, halogen atom, hydroxyl group, mercapto group, amino group, aldehyde group, carboxyl group, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, carbamoyl, nitro, cyano, amino, cyano, or a salt thereof,
R3Selected from hydrogen or-(CH)mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3,CO(CH2)3CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-6Alkyl, and, R312And R313Can form a quaternary, quinary or hexahydric aliphatic heterocyclic ring with nitrogen atoms;
R6is selected from
Figure BDA0002400750460000032
Wherein:
indicates the position of the bond
R61Represents one or more substituents independently selected from: hydrogen, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl, C1-6 alkoxy substituted C1-6 alkyl, carbamoyl, nitro, cyano, four-, five-or six-membered aliphatic ring or aliphatic heterocycle;
R7is selected from
Figure BDA0002400750460000041
Wherein:
R71optionally substituted or unsubstituted five-membered, six-membered aromatic ring or five-membered, six-membered aromatic heterocycle; the substituent on the substituted five-membered, six-membered aromatic ring or five-membered, six-membered aromatic heterocyclic ring may be one or more, and is arbitrarily selected from: : hydrogen, halogen atoms, hydroxy, mercaptoRadical, amino, aldehyde, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl, C1-6 alkoxy substituted C1-6 alkyl, carbamoyl, nitro, cyano.
g is an integer optionally selected from 0, 1,2,3 or 4, and represents CH when g is selected from 02Is absent;
h is an integer selected from 0, 1,2,3 or 4, and when h is selected from 0, it represents CH2Is absent;
y is optionally selected from carbon, nitrogen, oxygen, sulfur atoms or Y is absent.
Preferred R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-5Alkyl radical, C1-5Alkylene radical, C1-5Alkynyl, and, R312And R313Can form a five-membered or six-membered aliphatic heterocyclic ring with a nitrogen atom;
more preferred R31Independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-4Alkyl radical, C1-4Alkylene radical, C1-4Alkynyl, and, R312And R313Can form with nitrogen atoms
Figure BDA0002400750460000042
R314Represents one or more substituents independently selected from: hydrogen, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano,
Most preferred R32Is selected from COOCH3,COOCH2CH3
Most preferred R311Selected from hydroxy, methoxy, ethoxy, prop-2-alkynyl-1-amino
Most preferred R314Represents one or more substituents independently selected from: hydrogen, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl, methyl, ethyl, propyl, methoxyl, ethoxyl, propoxyl, methylmercapto, ethylsulfydryl, propylsulfydryl, trifluoromethyl, carbamyl, nitryl and cyano.
Preferred R61Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic or aliphatic heterocyclic ring;
more preferred R61Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic heterocycle;
further preferred R61Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl, methyl, ethyl, methoxyl, ethoxyl, methylmercapto, ethylsulfydryl, methylamino, ethylamino, dimethylaminoA diethylamino group,
Figure BDA0002400750460000051
Most preferred R61Represents one or more substituents independently selected from: hydrogen, methoxy, dimethylamino;
preferred R71Is selected from substituted or unsubstituted phenyl, and the substituent on the substituted phenyl can be one or more and is optionally selected from: : hydrogen, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl, C1-6 alkoxy substituted C1-6 alkyl, carbamoyl, nitro, cyano.
Preferably, g is an integer selected from 0, 1,2 or 3, and represents CH when g is selected from 02Is absent;
preferably, h is an integer arbitrarily selected from 0, 1,2 or 3, and when h is selected from 0, it represents CH2Is absent;
preferably Y is optionally absent from oxygen, sulfur atoms or Y;
preferred R711Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
More preferred R711Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
Most preferred R711Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl, methyl, ethyl and propylThe amino group comprises a group, a methoxy group, an ethoxy group, a propoxy group, a methylmercapto group, an ethylmercapto group, a propylmercapto group, a methylamino group, an ethylamino group, a propylamino group, a trifluoromethyl group, a carbamoyl group, a nitro group and a cyano group.
In a preferred embodiment of the invention, R is preferably61Is selected from
Figure BDA0002400750460000061
Or hydrogen;
in a preferred embodiment of the invention, R is preferably311And R312May be formed with nitrogen atoms and other carbon, nitrogen, oxygen, sulfur, etc. atoms
Figure BDA0002400750460000062
Figure BDA0002400750460000063
Preferred five-membered aryl groups of the present invention are selected from
Figure BDA0002400750460000064
Preferred six-membered aryl groups of the present invention are selected from
Figure BDA0002400750460000071
Preferred five-membered heteroaryl groups of the present invention are selected from:
Figure BDA0002400750460000072
preferred six membered heteroaryl groups of the present invention are selected from:
Figure BDA0002400750460000073
the four-element, five-element or six-element fat ring contains carbon atom saturated rings.
The quaternary aliphatic heterocyclic ring in the present invention includes, but is not limited to:
Figure BDA0002400750460000074
the five-membered aliphatic heterocyclic ring in the present invention includes, but is not limited to:
Figure BDA0002400750460000075
the six-membered aliphatic heterocyclic ring described in the present invention includes, but is not limited to:
Figure BDA0002400750460000076
if, in the present invention, the four-, five-, and six-membered aliphatic rings are not specifically mentioned, these aliphatic rings may be substituted or unsubstituted, and the substituent may be one or more substituents independently selected from: hydrogen, oxygen atom, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl radical, C1-6Alkoxy-substituted C1-6Alkyl, carbamoyl, nitro, cyano,
If the quaternary, quinary or hexabasic aliphatic ring is replaced by an oxygen atom, the oxygen atom and the carbon atom on the ring form a carbonyl group (C ═ O).
For example: the six-membered aliphatic ring described in the present invention includes, but is not limited to:
Figure BDA0002400750460000081
Figure BDA0002400750460000082
the quaternary, quinary or hexahydric aliphatic heterocyclic ring disclosed by the invention contains heteroatoms besides carbon atoms, wherein the heteroatoms comprise nitrogen atoms, oxygen atoms and sulfur atoms;
the quaternary aliphatic heterocyclic ring in the present invention includes, but is not limited to:
Figure BDA0002400750460000083
the five-membered aliphatic heterocyclic ring in the present invention includes, but is not limited to:
Figure BDA0002400750460000084
the six-membered aliphatic heterocyclic ring described in the present invention includes, but is not limited to:
Figure BDA0002400750460000085
if, in the present invention, the four-, five-, and six-membered aliphatic heterocyclic rings are not specifically mentioned, these aliphatic heterocyclic rings may be substituted or unsubstituted, and the substituent may be one or more substituents independently selected from: hydrogen, oxygen atom, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl radical, C1-6Alkoxy-substituted C1-6Alkyl, carbamoyl, nitro, cyano,
If the four-membered, five-membered and six-membered aliphatic heterocyclic ring is substituted by an oxygen atom, the oxygen atom and the carbon atom on the ring form a carbonyl group (C ═ O).
For example: the six-membered aliphatic heterocyclic ring described in the present invention includes, but is not limited to:
Figure BDA0002400750460000086
Figure BDA0002400750460000087
substituted or unsubstituted C in the invention1-6Substituents on the linear or branched alkyl groups are selected from: hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, carbamoyl, halogen, nitro, cyano;
c of the invention1-6Straight or branched alkyl, preferably C1-4Straight or branched alkyl or C2-5Straight or branched chain alkyl, more preferablyC2-4Straight or branched alkyl or C2-4Straight or branched alkyl, further preferred is C3-4Straight or branched alkyl or C2-3Straight or branched chain alkyl. More preferred is C1-6The linear or branched alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl, pentyl, isopentyl. More preferred is C1-4The linear or branched alkyl is selected from methyl, ethyl, propyl, isopropyl, butyl.
Preferred compounds of formula I include but are not limited to compounds of formula IA
Figure BDA0002400750460000091
R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3,CO(CH2)3CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-6Alkyl radical, C1-6Alkylene radical, C1-6Alkynyl, and, R312And R313Can form a quaternary, quinary or hexahydric aliphatic heterocyclic ring with nitrogen atoms;
R61represents one or more substituents independently selected from: hydrogen, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl radical, C1-6Alkoxy-substituted C1-6Alkyl, carbamoyl,Nitro, cyano, quaternary, quinary or hexahydric aliphatic rings or aliphatic heterocycles;
g is an integer selected from 0, 1,2 or 3, and when g is selected from 0, it represents CH2Is absent;
h is an integer selected from 0, 1,2 or 3, and when h is selected from 0, it represents CH2Is absent;
y is optionally selected from oxygen, sulfur atoms or Y is absent;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula I include, but are not limited to, compounds of formula IB
Figure BDA0002400750460000101
R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3,CO(CH2)3CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-6Alkyl radical, C1-6Alkylene radical, C1-6Alkynyl, and, R312And R313Can form a quaternary, quinary or hexahydric aliphatic heterocyclic ring with nitrogen atoms;
R61represents one or more substituents independently selected from: hydrogen, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl, C1-6 alkoxy substituted C1-6 alkyl, carbamoyl, nitro, cyano, four-, five-or six-membered aliphatic ring or aliphatic heterocycle;
g is an integer selected from 0, 1,2 or 3, and when g is selected from 0, it represents CH2Is absent;
h is an integer selected from 0, 1,2 or 3, and when h is selected from 0, it represents CH2Is absent;
y is optionally selected from oxygen, sulfur atoms or Y is absent;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula IA include, but are not limited to, compounds of formula IA1
Figure BDA0002400750460000111
R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3,CO(CH2)3CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-6Alkyl radical, C1-6Alkylene radical, C1-6Alkynyl, and, R312And R313Can form a quaternary, quinary or hexahydric aliphatic heterocyclic ring with nitrogen atoms;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic heterocycle;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula IA include, but are not limited to, compounds of formula IA2
Figure BDA0002400750460000112
Figure BDA0002400750460000121
Wherein the content of the first and second substances,
R311selected from hydrogen atoms, C1-6Alkyl radical, C1-6Alkylene radical, C1-6An alkynyl group;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, amino, and amino,Nitro, cyano, six-membered aliphatic heterocycle;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula IA2 include, but are limited to, compounds of formula IA2a
Figure BDA0002400750460000122
Wherein the content of the first and second substances,
R311independently selected from hydrogen atom, C1-4Alkyl radical, C1-4Alkylene radical, C1-4An alkynyl group;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl, methyl, ethyl, propyl, methoxyl, ethoxyl, propoxyl, methylmercapto, ethylsulfydryl, propylsulfydryl, methylamino, ethylamino, propylamino, trifluoromethyl, carbamoyl, nitro and cyano.
Preferred compounds of formula IA include, but are not limited to, compounds of formula IA3
Figure BDA0002400750460000123
Figure BDA0002400750460000131
R311Independently selected from hydrogen atom, C1-6Alkyl radical, C1-6Alkylene radical, C1-6An alkynyl group;
R32is selected from COOCH3,COOCH2CH3
R61Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl, methyl, ethyl, methoxyl, ethoxyl, methylmercapto, ethylsulfhydryl, methylamino, ethylamino, dimethylamino, diethylamino,
Figure BDA0002400750460000132
R711Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, methyl, ethyl, propyl, methoxyl, ethoxyl, propoxy, methylmercapto, ethylsulfhydryl, propylsulfhydryl, methylamino, ethylamino, propylamino, CF3Carbamoyl, nitro, cyano.
Preferred compounds of formula IA include, but are not limited to, compounds of formula IA4
Figure BDA0002400750460000133
R314Represents one or more substituents independently selected from: hydrogen, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic heterocycle;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula IA4 include, but are limited to, compounds of formula IA4a
Figure BDA0002400750460000141
R314Represents one or more substituents independently selected from: hydrogen, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl, methyl, ethyl, propyl, methoxyl, ethoxyl, propoxyl, methylmercapto, ethylsulfydryl, propylsulfydryl, trifluoromethyl, carbamyl, nitryl and cyano;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl, methyl, ethyl, propyl, methoxyl, ethoxyl, propoxyl, methylmercapto, ethylsulfydryl, propylsulfydryl, methylamino, ethylamino, propylamino, trifluoromethyl, carbamoyl, nitro and cyano.
Preferred compounds of formula IB include, but are limited to, compounds of formula IB1
Figure BDA0002400750460000142
R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-5Alkyl radical, C1-5Alkylene radical, C1-5Alkynyl, and, R312And R313Can form a five-membered or six-membered aliphatic heterocyclic ring with a nitrogen atom;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic heterocycle;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula IB include, but are limited to, compounds of formula IB2
Figure BDA0002400750460000151
R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atomsSeed, C1-5Alkyl radical, C1-5Alkylene radical, C1-5Alkynyl, and, R312And R313Can form a five-membered or six-membered aliphatic heterocyclic ring with a nitrogen atom;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic heterocycle;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula IB2 include, but are limited to, compounds of formula IB2a
Figure BDA0002400750460000161
Wherein R is311Selected from hydrogen atoms, C1-4Alkyl radical, C1-4Alkylene radical, C1-4An alkynyl group;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl, methyl, ethyl, methoxyl, ethoxyl, methylmercapto, ethylsulfhydryl, methylamino, ethylamino, dimethylamino, diethylamino,
Figure BDA0002400750460000162
R711Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, methyl, ethyl, propyl, methoxyl, ethoxyl, propoxyl, methylmercapto, and ethylideneMercapto, propylmercapto, methylamino, ethylamino, propylamino, CF3Carbamoyl, nitro, cyano.
Preferred compounds of formula IB2 include, but are limited to, compounds of formula IB2b
Figure BDA0002400750460000163
R314Represents one or more substituents independently selected from: hydrogen, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl, methyl, ethyl, methoxyl, ethoxyl, methylmercapto, ethylsulfhydryl, methylamino, ethylamino, dimethylamino, diethylamino,
Figure BDA0002400750460000171
R711Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, methyl, ethyl, propyl, methoxyl, ethoxyl, propoxy, methylmercapto, ethylsulfhydryl, propylsulfhydryl, methylamino, ethylamino, propylamino, CF3Carbamoyl, nitro, cyano.
Preferred compounds of formula IB include, but are limited to, compounds of formula IB3
Figure BDA0002400750460000172
R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-5Alkyl radical, C1-5Alkylene radical, C1-5Alkynyl, and, R312And R313Can form a five-membered or six-membered aliphatic heterocyclic ring with a nitrogen atom;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic heterocycle;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula II include but are limited to compounds of formula IIA
Figure BDA0002400750460000181
R61Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4An alkylamino group,Halogen substituted C1-4Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic or aliphatic heterocyclic ring;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula II include, but are not limited to, compounds of formula IIB
Figure BDA0002400750460000182
R11Represents one or more substituents independently selected from: hydrogen atom, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic or aliphatic heterocyclic ring;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula II include, but are not limited to, compounds of formula IIC
Figure BDA0002400750460000191
R11Represents one or more substituents independently selected from: hydrogen atom, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic or aliphatic heterocyclic ring;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula II include, but are not limited to, compounds of formula IID
Figure BDA0002400750460000192
Figure BDA0002400750460000201
R11Represents one or more substituents independently selected from: hydrogen atom, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3An alkylamino group,Halogen substituted C1-3Alkyl, carbamoyl, nitro, cyano.
R61Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic or aliphatic heterocyclic ring;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula II include, but are not limited to, compounds of formula IIE
Figure BDA0002400750460000202
R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-5Alkyl radical, C1-5Alkylene radical, C1-5Alkynyl, and, R312And R313Can form a five-membered or six-membered aliphatic heterocyclic ring with a nitrogen atom;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic or aliphatic heterocyclic ring;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
Preferred compounds of formula IIE include, but are limited to, compounds of formula IIE1
Figure BDA0002400750460000211
R311Independently selected from hydrogen atom, C1-4Alkyl radical, C1-4Alkylene radical, C1-4An alkynyl group;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic heterocycle;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
Most preferred compounds of formula (I), (II), stereoisomers, and pharmaceutically acceptable salts thereof, are selected from the group consisting of:
dimethyl (R) -2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) glutarate (Compound 1)
Dimethyl (R) -2- (7- (2- ((2-chloro-4-fluorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 2)
5-Ethyl-1-methyl (R) -2- (7- (2- ((2-chloro-4-fluorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 3)
Dimethyl (R) -2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 4)
5-Ethyl-1-methyl (R) -2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 5)
Dimethyl (R) -2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 6)
5-Ethyl-1-methyl (R) -2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 7)
Dimethyl (R) -2- (7- (2- ((3, 4-dichlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 8)
5-Ethyl-1-methyl (R) -2- (7- (2- ((3, 4-dichlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 9)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 10)
Methyl 2- (7- (2- (Benzyloxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 11)
Methyl 2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 12)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -7- (2- ((4-fluorobenzyl) oxa) acetoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 13)
Methyl 2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 14)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -7- (4- (4-fluorophenoxy) butanamide) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 15)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (Compound 16)
Methyl 2- (7- (4- (4-cyanophenoxy) butanamide) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 17)
Methyl 2- (7- (4-cyanobenzoylamino) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 18)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (4- (trifluoromethyl) benzoylamino) quinazolin-3 (4H) -yl) acetate (Compound 19)
N- (3- (2- (3, 3-Difluoropiperidin-1-yl) -2-oxoethyl) -6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 20)
N- (3- (2- (4, 4-Difluoropiperidin-1-yl) -2-oxoethyl) -6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 21)
N- (6- (3- (dimethylamino) phenoxy) -3- (2- (4-fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 22)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (piperidin-1-yl) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 23)
N- (6- (3- (dimethylamino) phenoxy) -3- (2- (4-fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4-fluorophenoxy) butanamide (compound 24)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 25)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4- (trifluoromethyl) phenoxy) butanamide (Compound 26)
2- ((4-cyanobenzyl) oxa) -N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) acetamide (Compound 27)
3-cyano-N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) benzamide (compound 28)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (trifluoromethyl) benzamide (compound 29)
Methyl 2- (6- (3-Morpholphenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 30)
N- (6- (3-Morpholphenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 31)
Methyl 2- (6- (2- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 32)
Methyl 2- (6- (4- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 33)
Methyl 2- (6- (4- (dimethylamino) phenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (compound 34)
Methyl 2- (7- (2- ((2, 4-dichlorobenzyl) oxa) acetoxy) -4-oxo-6-phenoxyquinazolin-3 (4H) -yl) acetate (Compound 35)
N- (6- (3-Morpholinophenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4- (trifluoromethyl) phenoxy) butanamide (Compound 36)
Methyl 2- (4-oxo-6-phenoxy-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 37)
Methyl 2- (4-oxo-6-phenoxy-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (Compound 38)
N- (3- (2- (4-Fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-6-phenoxy-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 39)
N- (3- (2- (3, 3-Difluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-6-phenoxy-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 40)
Methyl 2- (6- (3-morpholinophenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (Compound 41)
N- (3- (2- (3, 3-Difluoropiperidin-1-yl) -2-oxoethyl) -6- (3-morpholineoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 42)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) -1, 4-dihydroquinazolin-3 (2H) -yl) acetate (Compound 43)
N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclohexane-1, 2' -quinazolin-7 ' -yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 44)
N- (7- (3- (dimethylamino) phenoxy) -3 a-methyl-1, 5-dioxo-1, 2,3,3a,4, 5-hexahydropyrrolo [1,2-a ] quinazolin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 45)
N- (7- (3- (dimethylamino) phenoxy) -3 a-methyl-5-oxo-1, 2,3,3a,4, 5-hexahydropyrrolo [1,2-a ] quinazolin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 46)
2- ((4-cyanobenzyl) oxa) -N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclopentane-1, 2' -quinazolin-7 ' -yl) acetamide (Compound 47)
N- (6- (3- (dimethylamino) phenoxy) -2, 2-dimethyl-4-oxo-1, 2,3, 4-tetrahydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 48)
N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclopentane-1, 2' -quinazolin-7 ' -yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 49)
4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline e-7-amide (Compound 50)
Methyl 2- (6- (3-chlorophenyl) -4-oxo-7- ((2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (Compound 51)
6- (3-chlorophenyl) -4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -3, 4-dihydroquinazoline e-7-amide (Compound 52)
N- (3- (4-chlorophenyl) propyl) -6- (3-fluorophenyl) -4-oxo-3, 4-dihydroquinazoline e-7-amide (Compound 53)
6- (3-fluorophenyl) -4-oxo-N- (3-phenylpropyl) -3, 4-dihydroquinazoline e-7-amide (compound 54)
6- (3-chlorophenyl) -4-oxo-N- (3-phenylpropyl) -3, 4-dihydroquinazoline e-7-amide (Compound 55)
6- (3-chlorophenyl) -N- (3- (4-chlorophenyl) propyl) -4-oxo-3, 4-dihydroquinazoline e-7-amide (Compound 56)
4-oxo-N- (3-phenylpropyl) -6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline e-7-amide (compound 57)
N- (3- (4-chlorophenyl) propyl) -4-oxo-6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline e-7-amide (compound 58)
6- (3-fluorophenyl) -4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -3, 4-dihydroquinazoline e-7-amide (compound 59)
Methyl 2- (6- (3-chlorophenyl) -7- ((3- (4-chlorophenyl) propyl) carbamoyl) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 60)
Methyl 2- (6- (3-fluorophenyl) -4-oxo-7- ((2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (Compound 61)
Methyl 2- (6- (3-fluorophenyl) -4-oxo-7- ((3-phenylpropyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (Compound 62)
Methyl 2- (7- ((3- (4-chlorophenyl) propyl) carbamoyl) -6- (3-fluorophenyl) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 63)
Methyl 2- (6- (3-chlorophenyl) -4-oxo-7- ((3- (4- (trifluoromethyl) phenoxy) propyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (Compound 64)
6- (3-chlorophenyl) -4-oxo-N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (Compound 65)
2- (6- (3-chlorophenyl) -4-oxo-7- ((3- (4- (trifluoromethyl) phenoxy) propyl) carbamoyl) quinazolin-3 (4H) -yl) acetic acid (Compound 66)
6- (3-chlorophenyl) -4-oxo-3- (2-oxo-2- (piperidin-1-yl) ethyl) -N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (Compound 67)
6- (3-chlorophenyl) -3- (2- (3, 3-difluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (compound 68)
N- (6- (4- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 69)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 70)
N- (6- (3-Morpholphenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 71)
Figure BDA0002400750460000271
Figure BDA0002400750460000281
Figure BDA0002400750460000291
Figure BDA0002400750460000301
Figure BDA0002400750460000311
Figure BDA0002400750460000321
Figure BDA0002400750460000331
The second aspect of the invention also relates to processes for the preparation of the compounds of the invention.
Preparation method 1.
Figure BDA0002400750460000332
The synthetic reagent and reaction conditions used are (i) KNO3,H2SO480-130 ℃ for 6 h; (ii) phenol derivatives, NaHCO3,H2O,110℃,2h;(iii)HCl·H-L-Gly-OMe,HCl·D-Glu(OMe)-OMe or NH3·H2O, N, N' -Diisopropylcarbodiimide (DIC), THF, r.t. (iv) Fe, HCOOH,100 ℃,5 h; (v) EtOH, concentrated HCl; (vi)2- ((4- (trifluoromethyl) benzyl) oxa) acetyl chloride, Et3N,THF;(vii)LiOH,THF/H2O,1 h; (viii) HOSU, DIC; THF or DMF; r.t.,12 h; an amine derivative; THF; r.t.,12 h; (ix) formalin solution, NaBH3CN, AcOH, rt, overnight.
Preparation method 2.
Figure BDA0002400750460000341
The synthetic reagent and reaction conditions are (i) N-iodosuccinimide, HOAc, r.t.,24 h; (ii) phenylboronic acid derivatives, Pd (PPh)3)4,Na2CO3,80℃;(iii)LiOH,THF/H2O,1h;(iv)HCONH2160 ℃ for 5 hours; (v) triphosgene, THF, reflux; (vi) HCl-H-L-Gly-OMe, Et3N,H2O, r.t.; (vii) HCOOH,110 ℃,5 h; (viii) amine derivatives, HATU; (ix) HOSU, DIC, amine derivatives, THF; r.t.,12h
The third aspect of the present invention also relates to a pharmaceutical composition containing the compound of the present invention as an active ingredient. The pharmaceutical composition may be prepared according to methods well known in the art. The compounds of the invention may be formulated into any dosage form suitable for human or animal use by combining them with one or more pharmaceutically acceptable solid or liquid excipients and/or adjuvants. The compounds of the present invention are generally present in the pharmaceutical compositions in an amount of from 0.1 to 95% by weight.
The compounds of the present invention or pharmaceutical compositions containing them may be administered in unit dosage form by enteral or parenteral routes, such as oral, intravenous, intramuscular, subcutaneous, nasal, oromucosal, ophthalmic, pulmonary and respiratory, dermal, vaginal, rectal, and the like.
The dosage form for administration may be a liquid dosage form, a solid dosage form, or a semi-solid dosage form. The liquid dosage forms can be solution (including true solution and colloidal solution), emulsion (including o/w type, w/o type and multiple emulsion), suspension, injection (including water injection, powder injection and infusion), eye drop, nose drop, lotion, liniment, etc.; the solid dosage form can be tablet (including common tablet, enteric coated tablet, buccal tablet, dispersible tablet, chewable tablet, effervescent tablet, orally disintegrating tablet), capsule (including hard capsule, soft capsule, and enteric coated capsule), granule, powder, pellet, dripping pill, suppository, pellicle, patch, aerosol (powder), spray, etc.; semisolid dosage forms can be ointments, gels, pastes, and the like.
The compound can be prepared into common preparations, sustained release preparations, controlled release preparations, targeting preparations and various particle drug delivery systems.
For tableting the compounds of the invention, a wide variety of excipients known in the art may be used, including diluents, binders, wetting agents, disintegrants, lubricants, glidants. The diluent can be starch, dextrin, sucrose, glucose, lactose, mannitol, sorbitol, xylitol, microcrystalline cellulose, calcium sulfate, calcium hydrogen phosphate, calcium carbonate, etc.; the humectant can be water, ethanol, isopropanol, etc.; the binder can be starch slurry, dextrin, syrup, Mel, glucose solution, microcrystalline cellulose, acacia slurry, gelatin slurry, sodium carboxymethylcellulose, methylcellulose, hydroxypropyl methylcellulose, ethyl cellulose, acrylic resin, carbomer, polyvinylpyrrolidone, polyethylene glycol, etc.; the disintegrant may be dry starch, microcrystalline cellulose, low-substituted hydroxypropyl cellulose, crosslinked polyvinylpyrrolidone, crosslinked sodium carboxymethylcellulose, sodium carboxymethyl starch, sodium bicarbonate and citric acid, polyoxyethylene sorbitol fatty acid ester, sodium dodecyl sulfate, etc.; the lubricant and glidant may be talc, silicon dioxide, stearate, tartaric acid, liquid paraffin, polyethylene glycol, and the like.
The tablets may be further formulated into coated tablets, such as sugar-coated tablets, film-coated tablets, enteric-coated tablets, or double-layer and multi-layer tablets.
To encapsulate the administration units, the active ingredient of the compounds of the invention can be mixed with diluents and glidants and the mixture can be placed directly into hard or soft capsules. Or the effective component of the compound of the invention can be prepared into granules or pellets with diluent, adhesive and disintegrating agent, and then placed into hard capsules or soft capsules. The various diluents, binders, wetting agents, disintegrants, glidants used to prepare the compound tablets of the present invention may also be used to prepare capsules of the compound of the present invention.
In order to prepare the compound of the invention into injection, water, ethanol, isopropanol, propylene glycol or a mixture thereof can be used as a solvent, and a proper amount of solubilizer, cosolvent, pH regulator and osmotic pressure regulator which are commonly used in the field can be added. The solubilizer or cosolvent can be poloxamer, lecithin, hydroxypropyl-beta-cyclodextrin, etc.; the pH regulator can be phosphate, acetate, hydrochloric acid, sodium hydroxide, etc.; the osmotic pressure regulator can be sodium chloride, mannitol, glucose, phosphate, acetate, etc. For example, mannitol and glucose can be added as proppant for preparing lyophilized powder for injection.
In addition, colorants, preservatives, flavors, or other additives may also be added to the pharmaceutical preparation, if desired.
According to the fourth aspect of the invention, the compounds of the invention are used for carrying out biological activity screening on an intracellular pattern recognition receptor NOD1/2, and the results show that the quinazolinone compounds can remarkably antagonize NOD1/2 signal pathways, so that the compounds of the invention can be used for preparing medicines for preventing or treating immune inflammatory diseases and related diseases, especially for preparing anti-tumor medicines. The tumor includes but is not limited to melanoma, oral epidermoid carcinoma, esophageal carcinoma, gastric cancer, lung cancer, breast cancer, renal cancer, liver cancer, cervical cancer, ovarian cancer, pancreatic cancer, prostate cancer, colon cancer and bladder cancer.
For the purpose of administration and enhancing the therapeutic effect, the drug or pharmaceutical composition of the present invention can be administered by any known administration method.
The dosage of the pharmaceutical composition of the compound of the present invention to be administered may vary widely depending on the nature and severity of the disease to be prevented or treated, the individual condition of the patient or animal, the route and dosage form of administration, and the like. Generally, a suitable daily dosage range for a compound of the invention is from 0.001 to 150mg/Kg body weight, preferably from 0.1 to 100mg/Kg body weight, more preferably from 1 to 60mg/Kg body weight, and most preferably from 2 to 30mg/Kg body weight. The above-described dosage may be administered in one dosage unit or divided into several dosage units, depending on the clinical experience of the physician and the dosage regimen including the use of other therapeutic means.
The compounds or compositions of the present invention may be administered alone or in combination with other therapeutic or symptomatic agents. When the compound of the present invention is used in a synergistic manner with other therapeutic agents, the dosage thereof should be adjusted according to the actual circumstances.
The beneficial technical effects are as follows:
in the invention, through framework transition, quinazolinone is selected as a basic framework, and D-Gln or glycine is introduced to construct a novel compound library.
The quinazolinone compound can remarkably antagonize NOD1/2 signal channel, and is useful for preparing medicines for preventing or treating immunoinflammatory diseases and related diseases, especially for preparing antitumor medicines.
Description of the drawings:
FIG. 1, MDP and its analogs.
Figure 2 chemical structure of MDP derivatives.
FIG. 3 shows the lead compound 4h structure and NOD1/2 dual antagonistic activity.
FIG. 4, Compound 68 has significant antagonism of the hNOD1 and hNOD2 signaling pathways.
FIG. 5, compound 68 can significantly antagonize the agonistic action of C12-ie-DAP on NOD1 and NOD2 signaling pathways of THP1 cells, and reduce the transcription of inflammatory cytokines IL-6, TNF alpha and IL-8 mRNA.
FIG. 6, Compound 68 blocks NOD1 and NOD2 mediated activation of NF-. kappa.B and MAPK signaling pathways.
FIG. 7, Effect of compound 68 on mouse B16-F10 melanoma model tumor growth.
Detailed Description
The above-described aspects of the invention are explained in further detail below by means of specific embodiments in the form of chemical and biological examples. The various aspects and features of the present invention are illustrated in detail by the preferred examples of synthesis of the compounds of the present invention and the pharmacological test examples of the compounds of the present invention. It will be appreciated by those skilled in the art that these examples are for illustrative purposes only and should not be construed as limiting the scope of the above-described subject matter of the present invention to only the following examples. Various modifications and improvements to the various aspects of the invention may be made by those skilled in the art without departing from the scope of the claims and the specification, and these modifications and improvements are within the scope of the invention.
In addition, it should be noted that, unless otherwise specified, various materials and reagents used in the following examples are those commonly used in the art and are commercially available in a usual manner; the intermediates used may be obtained by conventional commercial routes or prepared by well-known methods; the methods used are all conventional methods known to the person skilled in the art.
[ chemical examples ]
Reagents for the experiments were purchased from Beijing coupling technologies, Inc., Bailingwei technologies, Inc., Acros Organics, Alfa Aesar, Sigma-Aldrich, and TCI, unless otherwise specified, and used without purification. The solvents used in the experiments were purchased mainly from Beijing chemical plant and Shigaku chemical Co., Ltd, and were used without further treatment except for THF and DMF which were further treated by the solvent purification system of VAC corporation. GF254 thin layer chromatography silica gel plate, GF254 silica gel thick preparation plate and silica gel powder (60-100 mesh, 160-.
HPLC-MS analyzer: the HPLC analyzer was an Agilent 1100HPLC system, an Agilent G1312A pump, an Agilent G1314A UV detector, an Agilent G1313A autosampler, an Agilent G1316A column oven and a diverter valve. The column was a Kromasil C18 analytical column (4.6 μm,4.6 mm. times.50 mm) available from two KMA companies. The mobile phase was acetonitrile containing 0.05% HCOOH and water. The linear gradient eluted 5:95(v: v) acetonitrile-H2O to 95:5(v: v) acetonitrile-H2O for 5minutes at a flow rate of 1 mL/min. UV detection wavelength 254 nm. The ThermoFinnigan LCQ-Advantage mass spectrometer is used, 5% of eluent is shunted into the mass spectrometer, and an electrospray ion source (ESI) is adopted in a positive ion or negative ion scanning mode. The method is mainly used for reaction monitoring and primary determination of compound purity.
UPLC-MS analyzer: the Acquity UPLC-MS system of Waters corporation includes a binary solvent manager, a sample manager, a chromatography column manager, a PDA detector, and an SQ mass spectrometer. The chromatographic column is Acquisty of Waters corporation
Figure BDA0002400750460000371
BEH C18 column (1.7 μm,2.1 mm. times.50 mm). The mobile phase was acetonitrile containing 0.05% HCOOH and water. The linear gradient eluted 5:95(v: v) acetonitrile-H2O to 95:5(v: v) acetonitrile-H2O over a period of 3minutes at a flow rate of 0.3 mL/min. UV detection wavelength 254 nm. The SQ mass spectrometer adopts a positive ion or negative ion scanning mode and an electrospray ionization source (ESI). The method is mainly used for reaction monitoring and primary determination of compound purity.
HPLC analyzer: an Agilent 1260HPLC system, an Agilent G1311C quaternary pump, an Agilent G4212B ultraviolet detector, an Agilent G1367E high-performance autosampler, and an Agilent G1316A column oven. A chiral analytical column: DAICEL CHIRALPAK AD-H, 250X 4.6mm, 5. mu.M (manufactured by DAICEL, Daiiol, Japan). The mobile phase is n-hexane/isopropanol and is eluted at equal intervals. UV detection wavelength 254 nm. The method is mainly used for analyzing the optical purity of the target compound.
High resolution mass spectrometer: agilent LC/MSD TOF system. A chromatographic column: agilent ZORBAX SB-C18(Rapid resolution,3.5 μm, 2.1X 30 mm). Mobile phase: MeOH H2O75: 25(v: v), containing 5mmol/L formic acid, isocratically eluted. The time is 5min, and the flow rate is 0.40 mL/min. The mass spectrum detection adopts a positive ion scanning mode and an electrospray ionization source (ESI). The method is mainly used for determining the accurate molecular weight of a target compound.
Nuclear magnetic resonance apparatus: varian Mercury 300MHz,400MHz,500MHz,600MHz and Bruker Avance 400MHz, solvent CDCl3,DMSO-d6。
Melting point apparatus: yanaco micro melting point apparatus, OptiMelt melting point apparatus, were not calibrated.
Example 1
Synthesis of dimethyl (R) -2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) glutarate (1)
Figure BDA0002400750460000381
15-chloro-2, 4-dinitrobenzoic acid (1-a)
Figure BDA0002400750460000382
While stirring at room temperature, 3-chlorobenzoic acid (10.0g,63.9mmol) was dissolved in 120mL of concentrated sulfuric acid, and potassium nitrate (16.5g,163.2mmol) was added portionwise over 15 min. The reaction solution reacts for 30min at 80 ℃, 2h at 110 ℃ and 2h at 120 ℃ in sequence. The reaction solution was poured into 660g of ice water, and filtered, and the obtained white solid was recrystallized from a mixed solvent of ethanol and water to obtain 8.08g of pale yellow crystals with a yield of 50.2%.1H NMR(400MHz,DMSO-d6)δ14.21(brs,1H),8.85(s,1H),8.28(s,1H).13C NMR(100MHz,DMSO-d6)δ163.78,147.95,145.70,132.86,132.06,130.52,122.05.
5- (3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (1-b)
Figure BDA0002400750460000391
Compound 1-a (4.877g,19.78mmol) was added to 36mL of water, sodium bicarbonate (3.556g,42.33mmol,2.14eq) and 3, 5-dimethylphenol (2.872g,20.97mmol,1.06eq) were added, and the mixture was heated to 110 ℃ and reacted under reflux for 2 hours until the starting material disappeared. Cooling, cooling in ice-water bath, adjusting pH to-3-4, filtering, draining, and drying to obtain compound 1-b with yield of 60%.1H NMR(400MHz,DMSO-d6)δ8.79(s,1H),7.29(t,J=8.2Hz,1H),7.14(s,1H),6.68(dd,J=8.4,2.2Hz,1H),6.59-6.57(m,1H),6.48(dd,J=7.9,1.9Hz,1H),2.92(s,6H).
(R) -dimethyl-2- (5- (3- (dimethylamino) phenoxy) -2, 4-dinitrobenzamido) pentanedioate (1-c)
Figure BDA0002400750460000392
The compound 1-b (4.546g,13.1mmol), D-glutamic acid dimethyl ester hydrochloride (2.909g,13.75mmol, 1.05eq), N, N-diisopropylcarbodiimide DIC (3.06mL, 19.65mmol, 1.5eq) were sequentially added to 120mL of THF, stirred at room temperature for 4h, filtered, evaporated to dryness, added with dichloromethane, washed with waterWashing with saturated salt solution, drying, filtering, evaporating to dryness, and performing column chromatography to obtain compound 1-c with yield of 70-85%.1H NMR(400MHz,DMSO-d6)δ9.15(d,J=7.7Hz,1H),8.85(s,1H),7.32-7.28(m,1H),6.96(s,1H),6.69(dd,J=8.4,2.2Hz,1H),6.57-6.56(m,1H),6.47(dd,J=7.9,2.0Hz,1H),4.43(td,J=8.8,5.3Hz,1H),3.58(d,J=8.1Hz,6H),2.92(s,6H),2.41–2.37(m,2H),2.05(td,J=13.3,7.7Hz,1H),1.85(dt,J=14.3,8.3Hz,1H).
Dimethyl (R) -2- (7-amino-6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (1-d)
Figure BDA0002400750460000401
Compound 1-c (210mg,0.464mmol) was added to 10mL of formic acid, iron powder (520mg,9.38mmol,20eq) was added, heated to 90 ℃ and reacted for 1.5-2.5h with LC-MS detection to the main point of the desired product (about 75%). Cooling, adding 20ml of EDC, passing through kieselguhr, evaporating the filtrate to dryness, and performing column chromatography with DCM/MeOH. The resulting solid was dissolved in 5mL of ethanol, and 0.5mL of concentrated hydrochloric acid was added to the solution to react at room temperature for 12 hours. Adding a large amount of diethyl ether into the reaction solution, separating out solid, filtering and drying to obtain the compound 1-d with the yield of 30-40%.
2- ((4- (trifluoromethyl) benzyl) oxy) acetic acid (1-e)
Figure BDA0002400750460000402
Under the protection of argon, adding an ice-water bath, adding sodium hydrogen (880mg, 60 percent and 22mmol) into 30mL THF, adding compound p-trifluoromethyl benzyl alcohol (10mmol and 1eq) dropwise under the ice-water bath, and stirring for 1h under the ice-water bath. 2-Bromoacetic acid (1.11g,8mmol,0.8eq)/20mL THF solution was added dropwise, and the mixture was allowed to warm to room temperature naturally and stirred overnight. And (3) quenching the reaction with water by using methanol, extracting impurities from methyl tert-butyl ether, adjusting the pH of a water phase to be 2-3 by using 6N HCl, extracting by using ethyl acetate, drying, filtering and evaporating to dryness to obtain a compound 1-e with the yield of 60.3%.
The title Compound (1), Compound 1-e (0.2mmol, 2eq) was dissolved in 4mL of dichloromethane and added dropwise to grass under ice-water bathAnd (3) transferring acyl chloride (0.4mmol, 4eq) to room temperature for reaction for 1h, allowing the raw material to disappear (detecting by adding methanol), evaporating the solvent to dryness, dissolving in 1mL of THF to obtain a THF solution of an intermediate, and directly using 100% of the THF solution for the next reaction. The THF solution obtained above was mixed with 1-d (0.1mmol) and triethylamine (0.5mmol,5eq) in 2mL of THF and reacted for 1 h. Silica gel column chromatography gave compound 1 in 60.1% yield.1H NMR(400MHz,DMSO-d6)δ9.68(s,1H),8.65(s,1H),8.30(s,1H),7.57(s,4H),7.32(s,1H),7.26(t,J=8.1Hz,1H),6.65(d,J=8.3Hz,1H),6.53(s,1H),6.39(d,J=7.7Hz,1H),5.16(s,1H),4.76(s,2H),4.34(s,2H),3.63(s,3H),3.47(s,3H),2.90(s,6H),2.46–2.27(m,4H).
Example 2
Synthesis of dimethyl (R) -2- (7- (2- ((2-chloro-4-fluorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (2)
Figure BDA0002400750460000411
2- ((2-chloro-4-fluorophenyl) oxy) acetic acid (2-a)
Figure BDA0002400750460000412
P-fluoro-m-bromobenzyl alcohol and 2-bromoacetic acid were reacted according to the method for synthesizing the compound 1-e described in example 1 to obtain 2-a.
The title compound (2) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 1-d and 2-a in 55% yield.1H NMR(400MHz,DMSO-d6)δ9.62(s,1H),8.64(s,1H),8.29(s,1H),7.60-7.58(m,1H),7.42-7.41(m,1H),7.29–7.24(m,2H),7.08–7.06(m,1H),6.65-6.63(m,1H),6.49(s,1H),6.39-6.37(m,1H),5.16(s,1H),4.71(s,2H),4.34(s,2H),3.63(s,3H),3.46(s,3H),2.90(s,6H),2.42–2.31(m,4H).
Example 3
Synthesis of 5-ethyl-1-methyl (R) -2- (7- (2- ((2-chloro-4-fluorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (3)
Figure BDA0002400750460000413
The title compound (3) was prepared as by-product 3 in 20% yield according to the synthesis of compound 1 described in example 1, using 1-d and 2-a.1H NMR(400MHz,DMSO-d6)δ9.61(s,1H),8.64(s,1H),8.29(s,1H),7.58(dd,J=8.2,6.7Hz,1H),7.41(dd,J=8.7,2.1Hz,1H),7.29(s,1H),7.25(d,J=8.2Hz,1H),7.06(ddd,J=8.4,6.4,2.2Hz,1H),6.65–6.63(m,1H),6.49(s,1H),6.38(d,J=7.9Hz,1H),5.17(dd,J=7.8,4.9Hz,1H),4.71(s,2H),4.34(s,2H),3.90(q,J=7.0Hz,2H),3.63(s,3H),2.90(s,6H),2.42–2.24(m,4H),1.08(t,J=7.1Hz,3H).
Example 4
Synthesis of dimethyl (R) -2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (4)
Figure BDA0002400750460000421
2- ((4-cyanophenyl) oxy) acetic acid (4-a)
Figure BDA0002400750460000422
P-cyanobenzyl alcohol and 2-bromoacetic acid were reacted according to the method for synthesizing compound 1-e described in example 1 to give 4-a.
The title compound (4) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 1-d and 4-a, in 45% yield.1H NMR(400MHz,DMSO-d6)δ9.66(s,1H),8.64(s,1H),8.30(s,1H),7.68(d,J=8.0Hz,2H),7.55(d,J=8.0Hz,2H),7.30(s,1H),7.28-7.24(m,1H),6.66-6.64(m,1H),6.49(s,1H),6.38(d,J=7.7Hz,1H),5.19–5.16(m,1H),4.75(s,2H),4.33(s,2H),3.63(s,3H),3.46(s,3H),2.90(s,6H),2.45–2.27(m,4H).
Example 5
Synthesis of 5-ethyl-1-methyl (R) -2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (5)
Figure BDA0002400750460000431
The title compound (5) was prepared as by-product 5 in 15% yield according to the synthesis of compound 1 described in example 1, using 1-d and 4-a.1H NMR(400MHz,DMSO-d6)δ9.66(s,1H),8.63(s,1H),8.29(s,1H),7.69(d,J=8.1Hz,2H),7.55(d,J=8.0Hz,2H),7.30(s,1H),7.28-7.24(m,1H),6.65(dd,J=8.4,1.5Hz,1H),6.49(s,1H),6.38-6.36(m,1H),5.17(dd,J=8.3,4.9Hz,1H),4.75(s,2H),4.33(s,2H),3.90(q,J=7.1Hz,2H),3.63(s,3H),2.90(s,6H),2.40–2.26(m,4H),1.08(t,J=7.1Hz,3H).
Example 6
Synthesis of dimethyl (R) -2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (6)
Figure BDA0002400750460000432
2- ((4-chlorophenyl) oxy) acetic acid (6-a)
Figure BDA0002400750460000433
P-chlorobenzyl alcohol and 2-bromoacetic acid were reacted according to the method for synthesizing compound 1-e described in example 1 to give 6-a.
The title compound (6) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 1-d and 6-a, 62% yield.1H NMR(400MHz,DMSO-d6)δ9.63(s,1H),8.65(s,1H),8.29(s,1H),7.38(d,J=8.2Hz,2H),7.31(s,1H),7.29–7.25(m,3H),6.66–6.64(m,1H),6.52(s,1H),6.39(dd,J=7.9,1.3Hz,1H),5.16(dd,J=8.9,4.7Hz,1H),4.64(s,2H),4.28(s,2H),3.63(s,3H),3.47(s,3H),2.91(s,6H),2.43–2.27(m,4H).
Example 7
Synthesis of 5-ethyl-1-methyl (R) -2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (7)
Figure BDA0002400750460000441
The title compound (7) was prepared as by-product 7 in 20% yield according to the synthesis of compound 1 described in example 1, using 1-d and 6-a.1H NMR(400MHz,DMSO-d6)δ9.63(s,1H),8.64(s,1H),8.29(s,1H),7.38(d,J=8.3Hz,2H),7.30(s,1H),7.29-7.25(m,3H),6.66(dd,J=8.3,1.8Hz,1H),6.52(s,1H),6.39(dd,J=8.0,0.8Hz,1H),5.17(dd,J=8.1,5.0Hz,1H),4.64(s,2H),4.28(s,2H),3.90(q,J=7.0Hz,2H),3.63(s,3H),2.91(s,6H),2.40–2.26(m,4H),1.08(t,J=7.1Hz,3H).
Example 8
Synthesis of dimethyl (R) -2- (7- (2- ((3, 4-dichlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (8)
Figure BDA0002400750460000442
2- ((3, 4-dichlorophenyl) oxy) acetic acid (8-a)
Figure BDA0002400750460000451
3, 4-dichlorobenzyl alcohol and 2-bromoacetic acid were reacted according to the synthesis of compound 1-e described in example 1 to give 8-a.
The title compound (8) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 1-d and 8-a, 42% yield.1H NMR(400MHz,DMSO-d6)δ9.64(s,1H),8.64(s,1H),8.29(s,1H),7.64(d,J=1.4Hz,1H),7.48(d,J=8.2Hz,1H),7.36(dd,J=8.2,1.5Hz,1H),7.28(s,1H),7.25(d,J=8.2Hz,1H),6.64(dd,J=8.4,2.0Hz,1H),6.50(d,J=2.0Hz,1H),6.38(dd,J=7.8,1.7Hz,1H),5.16(dd,J=8.8,4.8Hz,1H),4.65(s,2H),4.31(s,2H),3.63(s,3H),3.46(s,3H),2.90(s,6H),2.43–2.28(m,4H).
Example 9
Synthesis of 5-ethyl-1-methyl (R) -2- (7- (2- ((3, 4-dichlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (9)
Figure BDA0002400750460000452
The title compound (9) was prepared as by-product 7 in 23% yield according to the synthesis of compound 1 described in example 1, using 1-d and 8-a.1H NMR(400MHz,DMSO-d6)δ9.65(s,1H),8.63(s,1H),8.29(s,1H),7.64(d,J=1.3Hz,1H),7.50-7.47(m,1H),7.36(dd,J=8.4,1.3Hz,1H),7.28(s,1H),7.24(d,J=8.2Hz,1H),6.64(dd,J=8.4,2.0Hz,1H),6.50(s,1H),6.38(dd,J=7.8,1.7Hz,1H),5.16(dd,J=8.8,4.6Hz,1H),4.66(s,2H),4.31(s,2H),3.89(q,J=7.1Hz,2H),3.63(s,3H),2.90(s,6H),2.42–2.24(m,4H),1.07(t,J=7.1Hz,3H).
Example 10
Synthesis of methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (10)
Figure BDA0002400750460000461
Methyl (5- (3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoyl) glycine (10-a)
Figure BDA0002400750460000462
Following the synthesis of compound 1-c described in example 1, 1-b was reacted with glycine methyl ester hydrochloride to give 10-a.
Methyl 2- (7-amino-6- (3- (dimethylamino) phenoxy) -4-oxaquinazolin-3 (4H) -yl) acetate (10-b)
Figure BDA0002400750460000463
According to the synthesis method of the compound 1-d in the embodiment 1, 10-a reacts with iron powder, and then reacts with hydrochloric acid after purification to obtain 10-d.
The title compound (10) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-bd and 1-e in 78% yield.1H NMR(400MHz,DMSO-d6)δ9.67(s,1H),8.66(s,1H),8.30(s,1H),7.57(s,4H),7.36(s,1H),7.27-7.23(m,1H),6.64(dd,J=8.4,1.9Hz,1H),6.53(s,1H),6.38(dd,J=7.9,1.5Hz,1H),4.78-4.76(m,4H),4.33(s,2H),3.68(s,3H),2.89(s,6H).
Example 11
Synthesis of methyl 2- (7- (2- (benzyloxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (11)
Figure BDA0002400750460000471
2- (benzyloxy) acetic acid (11-a)
Figure BDA0002400750460000472
Benzyl alcohol was reacted with 2-bromoacetic acid according to the synthesis of compound 1-e described in example 1 to give 11-a.
The title compound (11) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and 11-a in 78% yield.1H NMR(400MHz,DMSO-d6)δ9.63(s,1H),8.66(s,1H),8.30(s,1H),7.37–7.35(m,2H),7.33(s,1H),7.27(dd,J=10.1,6.2Hz,4H),6.65(dd,J=8.3,1.9Hz,1H),6.54(s,1H),6.41(dd,J=7.9,1.6Hz,1H),4.78(s,2H),4.65(s,2H),4.27(s,2H),3.68(s,3H),2.90(s,6H).
Example 12
Synthesis of methyl 2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (12)
Figure BDA0002400750460000473
The title compound (12) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and 4-a in 80% yield.1H NMR(400MHz,DMSO-d6)δ9.64(s,1H),8.64(s,1H),8.30(s,1H),7.68(d,J=8.1Hz,2H),7.55(d,J=8.1Hz,2H),7.35(s,1H),7.28-7.23(m,1H),6.64(dd,J=8.3,2.1Hz,1H),6.49-6.47m,1H),6.36(dd,J=7.9,1.8Hz,1H),4.78(s,2H),4.75(s,2H),4.33(s,2H),3.68(s,3H),2.90(s,6H).
Example 13
Synthesis of methyl 2- (6- (3- (dimethylamino) phenoxy) -7- (2- ((4-fluorobenzyl) oxa) acetoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (13)
Figure BDA0002400750460000481
2- ((4-Fluorobenzyl) oxy) acetic acid (13-a)
Figure BDA0002400750460000482
P-fluorobenzyl alcohol and 2-bromoacetic acid were reacted according to the synthesis of compound 1-e described in example 1 to give 13-a.
The title compound (13) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and 13-a in 85% yield.1H NMR(400MHz,DMSO-d6)δ9.62(s,1H),8.65(s,1H),8.29(s,1H),7.33(s,1H),7.31–7.24(m,2H),7.23-7.18(m,2H),7.14–7.05(m,1H),6.63(dd,J=8.3,2.1Hz,1H),6.53-6.4(m,1H),6.39(dd,J=7.9,1.9Hz,1H),4.78(s,2H),4.67(s,2H),4.29(s,2H),3.68(s,3H),2.90(s,6H).
Example 14
Synthesis of methyl 2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (14)
Figure BDA0002400750460000491
The title compound (14) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and 6-a in 83% yield.1H NMR(400MHz,DMSO-d6)δ9.62(s,1H),8.66(s,1H),8.30(s,1H),7.36(d,J=8.4Hz,3H),7.27(d,J=8.2Hz,3H),6.65(dd,J=8.4,2.1Hz,1H),6.52(t,J=2.2Hz,1H),6.38(dd,J=7.9,1.8Hz,1H),4.78(s,2H),4.64(s,2H),4.28(s,2H),3.68(s,3H),2.91(s,6H).
Example 15
Synthesis of methyl 2- (6- (3- (dimethylamino) phenoxy) -7- (4- (4-fluorophenoxy) butanamide) -4-oxoquinazolin-3 (4H) -yl) acetate (15)
Figure BDA0002400750460000492
4- (4-fluorophenoxy) butanoic acid (15-a)
Figure BDA0002400750460000493
According to the synthesis method of the compound 1-e described in example 1, p-fluorophenol and ethyl 4-bromobutyrate are reacted and subjected to column chromatography. Dissolving the solid in MeOH, adding an isovolumetric 1M sodium hydroxide solution, reacting overnight at room temperature until the raw materials completely disappear, adding 6N hydrochloric acid for neutralization, separating out white solid, filtering, and drying to obtain 15-a.
The title compound (15) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and 15-a in 80% yield.1H NMR(400MHz,DMSO-d6)δ9.90(s,1H),8.59(s,1H),8.26(s,1H),7.28–7.22(m,2H),7.08(dd,J=10.9,4.4Hz,2H),6.95–6.91(m,2H),6.62(dd,J=8.4,2.3Hz,1H),6.51-6.49(m,1H),6.37(dd,J=7.9,2.0Hz,1H),4.77(s,2H),3.99(t,J=6.3Hz,2H),3.67(s,3H),2.90(s,6H),2.70(t,J=7.3Hz,2H),2.06–2.01(m,2H).
Example 16
Synthesis of methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (16)
Figure BDA0002400750460000501
4- (4- (trifluoromethyl) phenoxy) butanoic acid (16-a)
Figure BDA0002400750460000502
P-trifluoromethylphenol and ethyl 4-bromobutyrate were reacted according to the method for synthesizing the compound 15-a described in example 15 to obtain 16-a.
The title compound (16) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and 16-a, in 82% yield.1H NMR(400MHz,DMSO-d6)δ9.92(s,1H),8.59(s,1H),8.26(s,1H),7.63(d,J=8.7Hz,2H),7.27(s,1H),7.27-7.21(m,1H),7.10(d,J=8.6Hz,2H),6.62(dd,J=8.3,2.2Hz,1H),6.51-6.48(m,1H),6.36(dd,J=7.8,1.9Hz,1H),4.77(s,2H),4.12(t,J=6.3Hz,2H),3.67(s,3H),2.90(s,6H),2.74–2.70(m,2H),2.07(dd,J=13.7,6.8Hz,2H).
Example 17
Synthesis of methyl 2- (7- (4- (4-cyanophenoxy) butanamide) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (17)
Figure BDA0002400750460000511
4- (4-Cyanophenoxy) butyl (17-a)
Figure BDA0002400750460000512
P-cyanophenol and ethyl 4-bromobutyrate were reacted according to the synthesis of compound 15-a described in example 15 to give 17-a.
The title compound (17) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and 17-a, 71% yield.1H NMR(400MHz,DMSO-d6)δ9.91(s,1H),8.59(s,1H),8.26(s,1H),7.74(d,J=8.8Hz,2H),7.27(s,1H),7.23(d,J=8.1Hz,1H),7.09(d,J=8.8Hz,2H),6.61(dd,J=8.4,2.1Hz,1H),6.49(t,J=2.1Hz,1H),6.36(dd,J=7.9,1.7Hz,1H),4.77(s,2H),4.13(t,J=6.3Hz,2H),3.67(s,3H),2.90(s,6H),2.72(t,J=7.2Hz,2H),2.08(dd,J=13.4,6.7Hz,2H).
Example 18
Synthesis of methyl 2- (7- (4-cyanobenzoylamino) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (18)
Figure BDA0002400750460000513
The title compound (18) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and p-cyanobenzoyl chloride in 78% yield.1H NMR(400MHz,DMSO-d6)δ10.36(s,1H),8.34(d,J=19.6Hz,2H),8.07(d,J=8.3Hz,2H),8.02(d,J=8.3Hz,2H),7.40(s,1H),7.26-7.21(m,1H),6.60(dd,J=8.3,2.0Hz,1H),6.51(s,1H),6.39(dd,J=7.9,1.5Hz,1H),4.80(s,2H),3.69(s,3H),2.89(s,6H).
Example 19
Synthesis of methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (4- (trifluoromethyl) benzoylamino) quinazolin-3 (4H) -yl) acetate (19)
Figure BDA0002400750460000521
The title compound (19) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 10-b and p-trifluoromethylbenzoyl chloride in 70% yield.1H NMR(400MHz,DMSO-d6)δ10.32(s,1H),8.35(d,J=28.7Hz,2H),8.11(d,J=8.0Hz,2H),7.91(d,J=8.2Hz,2H),7.40(s,1H),7.26-7.20(m,1H),6.60(dd,J=8.4,1.8Hz,1H),6.52(s,1H),6.40(d,J=7.9Hz,1H),4.80(s,2H),3.69(s,3H),2.88(s,6H).
Example 20
Synthesis of N- (3- (2- (3, 3-difluoropiperidin-1-yl) -2-oxoethyl) -6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (20)
Figure BDA0002400750460000522
The title compound (20) 10(1mmol) was dissolved in 6mL THF and 3mLH2And adding LiOH (3mmol,3eq) into the O mixed solution, reacting at room temperature for 0.5h till the reaction is complete, evaporating the reaction solution to dryness, adding water for dilution, adjusting the pH value to acidity by 1N hydrochloric acid, separating out a solid, filtering, and drying to obtain an intermediate product with the yield of 82%. The intermediate (0.5mmol), DIC (1mmol, 2eq) and HoSu (1mmol, 2eq) were dissolved in THF solution, stirred at room temperature overnight, added with 3, 3-difluoropiperidine hydrochloride (1.5mmol, 3eq) and reacted for 0.5h, the solvent was evaporated and separated by column chromatography to give compound 20 with 62% yield.1H NMR(400MHz,DMSO-d6)δ9.65(s,1H),8.65(s,1H),8.19(s,1H),7.56(s,4H),7.36(s,1H),7.24(t,J=8.0Hz,1H),6.63(d,J=7.8Hz,1H),6.52(s,1H),6.37(d,J=7.5Hz,1H),4.91(d,J=20.1Hz,2H),4.75(s,2H),4.33(s,2H),3.91(t,J=11.2Hz,1H),3.78(t,J=11.9Hz,1H),3.53(d,J=32.2Hz,2H),2.89(s,6H),2.12–2.08(m,2H),1.77(s,1H),1.61(s,1H).
Example 21
Synthesis of N- (3- (2- (4, 4-difluoropiperidin-1-yl) -2-oxoethyl) -6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (21)
Figure BDA0002400750460000531
The title compound (21) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 10 and 4, 4-difluoropiperidine hydrochloride in 46% yield.1H NMR(400MHz,DMSO-d6)δ9.65(s,1H),8.65(s,1H),8.18(s,1H),7.56(s,4H),7.37(s,1H),7.24(s,1H),6.64(s,1H),6.52(s,1H),6.37(s,1H),4.94(s,2H),4.75(s,2H),4.32(s,2H),3.60(d,J=31.1Hz,4H),2.88(s,6H),2.14–1.92(m,4H).
Example 22
Synthesis of N- (6- (3- (dimethylamino) phenoxy) -3- (2- (4-fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (22)
Figure BDA0002400750460000541
The title compound (22) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 10 and 4-fluoropiperidine hydrochloride in 50% yield.1H NMR(400MHz,DMSO-d6)δ9.65(s,1H),8.65(s,1H),8.18(s,1H),7.56(s,4H),7.36(s,1H),7.24(t,J=8.1Hz,1H),6.64–6.62(m,1H),6.52(s,1H),6.36(dd,J=7.8,1.1Hz,1H),4.90(d,J=3.8Hz,2H),4.75(s,2H),4.33(s,2H),3.67–3.61(m,1H),3.56–3.43(m,4H),2.89(s,6H),2.03–1.96(m,1H),1.80(dd,J=17.9,9.4Hz,2H),1.68–1.56(m,1H).
Example 23
Synthesis of N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (piperidin-1-yl) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (23)
Figure BDA0002400750460000542
The title compound (23) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 10 and piperidine in 45% yield.1H NMR(400MHz,DMSO-d6)δ9.64(s,1H),8.64(s,1H),8.18(s,1H),7.56(s,4H),7.36(s,1H),7.24(t,J=8.1Hz,1H),6.63(d,J=7.4Hz,1H),6.52(s,1H),6.36(d,J=7.6Hz,1H),4.85(s,2H),4.75(s,2H),4.33(s,2H),3.44(d,J=24.5Hz,4H),2.89(s,6H),1.59(s,4H),1.44(s,2H).
Example 24
Synthesis of N- (6- (3- (dimethylamino) phenoxy) -3- (2- (4-fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4-fluorophenoxy) butanamide (24)
Figure BDA0002400750460000551
The title compound (24) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 15 and 4-fluoropiperidine hydrochloride in 40% yield.1H NMR(400MHz,DMSO-d6)δ9.87(s,1H),8.57(s,1H),8.14(s,1H),7.28(s,1H),7.26-7.21(m,1H),7.10–7.06(m,2H),6.94–6.91(m,2H),6.61(dd,J=8.3,2.2Hz,1H),6.51-6.48(m,1H),6.36(dd,J=7.9,1.8Hz,1H),4.88(d,J=3.6Hz,2H),3.98(d,J=6.3Hz,2H),3.66–3.60(m,1H),3.48(dt,J=19.5,11.1Hz,4H),2.90(s,6H),2.70(t,J=7.3Hz,2H),2.05–2.01(m,2H),1.71(dt,J=29.8,14.7Hz,4H).
Example 25
Synthesis of N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (25)
Figure BDA0002400750460000552
5- (3- (dimethylamino) phenoxy) -2, 4-dinitrobenzamide (25-a)
Figure BDA0002400750460000553
1-b was reacted with aqueous ammonia according to the synthesis of compound 1-c described in example 1 to give 25-a.
7-amino-6- (3- (dimethylamino) phenoxy) quinazolin-4 (3H) -one (25-b)
Figure BDA0002400750460000561
According to the synthesis method of the compound 1-d described in example 1, 25-a is reacted with iron powder, purified and reacted with hydrochloric acid to obtain 25-b.
The title compound (2)5) Prepared according to the method for the synthesis of compound 1 described in example 1, using 25-b and 1-e, yield 72%.1H NMR(400MHz,DMSO-d6)δ12.20(br s,1H),9.62(s,1H),8.61(s,1H),8.03(s,1H),7.56(s,4H),7.35(s,1H),7.2-7.23(m,1H),6.63(dd,J=8.1,1.6Hz,1H),6.51(s,1H),6.37(d,J=8.1Hz,1H),4.75-4.72(m,2H),4.32-4.29(m,2H),2.89(s,6H).
Example 26
Synthesis of N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4- (trifluoromethyl) phenoxy) butanamide (26)
Figure BDA0002400750460000562
The title compound (26) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 25-b and 16-a, yield 70%.1H NMR(400MHz,DMSO-d6)δ12.11(s,1H),9.86(s,1H),8.53(s,1H),7.99(s,1H),7.62(d,J=8.7Hz,2H),7.27(s,1H),7.27-7.21(m,1H),7.10(d,J=8.6Hz,2H),6.61(dd,J=8.4,2.1Hz,1H),6.49(t,J=2.2Hz,1H),6.36(dd,J=7.9,1.8Hz,1H),4.11(t,J=6.3Hz,2H),2.90(s,6H),2.71(t,J=7.3Hz,2H),2.08(dd,J=13.4,6.7Hz,2H).
Example 27
Synthesis of 2- ((4-cyanobenzyl) oxa) -N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) acetamide (27)
Figure BDA0002400750460000571
The title compound (27) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 25-b and 4-a, 55% yield.1H NMR(400MHz,DMSO-d6)δ9.50(s,1H),8.51(s,1H),8.02(s,1H),7.68(d,J=8.2Hz,2H),7.52(d,J=8.2Hz,2H),7.34(s,1H),7.25-7.20(m,1H),6.60(dd,J=8.4,2.2Hz,1H),6.44(t,J=2.2Hz,1H),6.31(dd,J=8.0,1.9Hz,1H),4.72(s,2H),4.29(s,2H),2.89(s,6H).
Example 28
Synthesis of 3-cyano-N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) benzamide (28)
Figure BDA0002400750460000572
The title compound (28) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 25-b and m-cyanobenzoyl chloride in 50% yield.1H NMR(400MHz,DMSO-d6)δ12.21(s,1H),10.30(s,1H),8.28(d,J=5.8Hz,2H),8.20(d,J=7.9Hz,1H),8.10–8.03(m,2H),7.76-7.71(m,1H),7.42(s,1H),7.25-7.17(m,1H),6.58(d,J=8.4Hz,1H),6.50(s,1H),6.37(d,J=7.8Hz,1H),2.89(s,6H).
Example 29
Synthesis of N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (trifluoromethyl) benzamide (29)
Figure BDA0002400750460000581
The title compound (29) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 25-b and p-trifluoromethylbenzoyl chloride in 67% yield.1H NMR(400MHz,DMSO-d6)δ12.20(s,1H),10.28(s,1H),8.33(s,1H),8.10(d,J=8.1Hz,2H),8.04(s,1H),7.90(d,J=8.2Hz,2H),7.40(s,1H),7.26-7.21(m,1H),6.59(dd,J=8.4,2.0Hz,1H),6.52-6.50(m,1H),6.39(dd,J=7.9,1.6Hz,1H),2.88(s,6H).
Example 30
Synthesis of methyl 2- (6- (3-morpholinophenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (30)
Figure BDA0002400750460000582
5- (3-Morpholinphenoxy) -2, 4-dinitrobenzoic acid (30-a)
Figure BDA0002400750460000583
1-a and 3-morpholinophenol were reacted according to the synthesis method of compound 1-b described in example 1 to give 30-a.
Methyl (5- (3-morpholinophenoxy) -2, 4-dinitrobenzoyl) glycine (30-b)
Figure BDA0002400750460000591
30-a was reacted with glycine methyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to give 30-b.
Methyl 2- (7-amino-6- (3-morpholinophenoxy) -4-oxaquinazolin-3 (4H) -yl) acetate (30-c)
Figure BDA0002400750460000592
According to the synthesis method of the compound 1-d in the embodiment 1, 30-b reacts with iron powder, and 30-c is obtained after column chromatography and concentrated hydrochloric acid reaction.
The title compound (30) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 30-c and 1-e in 68% yield.1H NMR(400MHz,DMSO-d6)δ9.67(s,1H),8.66(s,1H),8.30(s,1H),7.61(d,J=8.6Hz,2H),7.58(s,2H),7.34(s,1H),7.33-7.28(m,1H),6.87(dd,J=8.3,2.1Hz,1H),6.82-6.79(m,1H),6.57(dd,J=8.0,2.0Hz,1H),4.77(d,J=11.5Hz,4H),4.33(d,J=3.4Hz,2H),3.77–3.68(m,8H),3.68(s,3H).
Example 31
Synthesis of N- (6- (3-morpholinophenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (31)
Figure BDA0002400750460000593
5-(3-morpholinophenoxy)-2,4-dinitrobenzamide(31-a)
Figure BDA0002400750460000601
According to the synthesis of compound 1-c described in example 1, 30-a was reacted with ammonia to give 31-a.
7-amino-6- (3-morpholinophenoxy) quinazolin-4 (3H) -one (31-b)
Figure BDA0002400750460000602
According to the synthesis method of the compound 1-d in the embodiment 1, 31-a reacts with iron powder, and the obtained product reacts with concentrated hydrochloric acid after column chromatography to obtain 31-b.
The title compound (31) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 31-b and 1-e in 60% yield.1H NMR(400MHz,DMSO-d6)δ12.19(s,1H),9.62(s,1H),8.61(s,1H),8.03(s,1H),7.57(s,4H),7.33(d,J=2.1Hz,1H),7.30(d,J=8.2Hz,1H),6.86(dd,J=8.3,2.0Hz,1H),6.81-6.78(m,1H),6.55(dd,J=7.9,1.9Hz,1H),4.75(s,2H),4.31(s,2H),3.72–3.69(m,4H),3.13–3.08(m,4H).
Example 32
Synthesis of methyl 2- (6- (2- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (32)
Figure BDA0002400750460000611
5- (2- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (32-a)
Figure BDA0002400750460000612
Following the synthesis of compound 1-b described in example 1, 1-a was reacted with a bis-phenol to give 32-a.
Methyl (5- (2- (dimethylamino) phenoxy) -2, 4-dinitrobenzoyl) glycine (32-b)
Figure BDA0002400750460000613
32-a was reacted with glycine methyl ester hydrochloride according to the synthesis of compound 1-c described in example 1 to give 32-b.
Methyl 2- (7-amino-6- (2- (dimethylamino) phenoxy) -4-oxaquinazolin-3 (4H) -yl) acetate (32-c)
Figure BDA0002400750460000614
According to the synthesis method of the compound 1-d in the embodiment 1, 32-b reacts with iron powder, and 32-c is obtained after column chromatography and concentrated hydrochloric acid reaction.
The title compound (32) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 32-c and 1-e in 59% yield.1H NMR(400MHz,DMSO-d6)δ9.77(s,1H),8.67(s,1H),8.29(s,1H),7.57(d,J=6.7Hz,4H),7.26(dd,J=11.2,4.1Hz,1H),7.12(dd,J=8.1,1.4Hz,2H),7.07(s,1H),7.03(dd,J=10.8,4.4Hz,1H),4.77(d,J=5.4Hz,4H),4.37(s,2H),3.67(s,3H),2.61(s,6H).
Example 33
Synthesis of methyl 2- (6- (4- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (33)
Figure BDA0002400750460000621
5- (4- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (33-a)
Figure BDA0002400750460000622
1-a was reacted with p-dimethylaminophenol according to the method for synthesizing compound 1-b described in example 1 to give 33-a.
Methyl (5- (4- (dimethylamino) phenoxy) -2, 4-dinitrobenzoyl) glycine (33-b)
Figure BDA0002400750460000623
33-a was reacted with glycine methyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to give 33-b.
Methyl 2- (7-amino-6- (4- (dimethylamino) phenoxy) -4-oxaquinazolin-3 (4H) -yl) acetate (33-c)
Figure BDA0002400750460000631
According to the synthesis method of the compound 1-d in the embodiment 1, 33-b reacts with iron powder, and after column chromatography, the compound reacts with concentrated hydrochloric acid to obtain 33-c.
The title compound (33) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 33-c and 1-e in 52% yield.1H NMR(400MHz,DMSO-d6)δ9.71(s,1H),8.64(s,1H),8.28(s,1H),7.58(d,J=9.1Hz,4H),7.22(s,1H),7.06(d,J=8.4Hz,2H),6.81(d,J=8.4Hz,2H),4.77(s,4H),4.34(s,2H),3.67(s,3H),2.92(s,6H).
Example 34
Synthesis of methyl 2- (6- (4- (dimethylamino) phenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (34)
Figure BDA0002400750460000632
The title compound (34) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 33-c and 16-a, 54% yield.1H NMR(400MHz,DMSO-d6)δ9.93(s,1H),8.60(s,1H),8.25(s,1H),7.63(d,J=8.6Hz,2H),7.14(s,1H),7.11(d,J=8.5Hz,2H),7.03(d,J=9.0Hz,2H),6.80(d,J=9.0Hz,2H),4.75(s,2H),4.13(t,J=6.3Hz,2H),3.67(s,3H),2.92(s,6H),2.75(t,J=7.2Hz,2H),2.13-2.04(m,2H).
Example 35
Synthesis of methyl 2- (7- (2- ((2, 4-dichlorobenzyl) oxa) acetoxy) -4-oxo-6-phenoxyquinazolin-3 (4H) -yl) acetate (35)
Figure BDA0002400750460000641
2, 4-dinitro-5-phenoxybenzoic acid (35-a)
Figure BDA0002400750460000642
1-a was reacted with phenol according to the synthesis method of compound 1-b described in example 1 to give 35-a.
Methyl (2, 4-dinitro-5-phenoxybenzoyl) glycine (35-b)
Figure BDA0002400750460000643
35-a was reacted with glycine methyl ester hydrochloride according to the synthesis of compound 1-c described in example 1 to give 35-b.
Methyl 2- (7-amino-4-oxa-6-phenoxyquinazolin-3 (4H) -yl) acetate (35-c)
Figure BDA0002400750460000644
According to the synthesis method of the compound 1-d in the embodiment 1, 35-b reacts with iron powder, and after column chromatography, the reaction is carried out with concentrated hydrochloric acid to obtain 35-c.
2- ((2, 4-dichlorobenzyl) oxy) acetic acid (35-d)
Figure BDA0002400750460000651
2, 4-dichlorobenzyl alcohol and 2-bromoacetic acid were reacted according to the synthesis method of compound 1-e described in example 1 to give 35-d.
The title compound (35) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 35-c and 35-d in 88% yield.1H NMR(400MHz,DMSO-d6)δ9.65(s,1H),8.66(s,1H),8.32(s,1H),7.59(s,1H),7.54(d,J=8.3Hz,1H),7.51-7.46(m,2H),7.31(s,1H),7.28(d,J=10.8Hz,2H),7.17(d,J=7.9Hz,2H),4.74(d,J=32.1Hz,4H),4.35(s,2H),3.68(s,3H).
Example 36
Synthesis of N- (6- (3-morpholinophenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4- (trifluoromethyl) phenoxy) butanamide (36)
Figure BDA0002400750460000652
The title compound (36) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 31-b and 16-a, in 88% yield.1H NMR(400MHz,DMSO-d6)δ12.12(s,1H),9.86(s,1H),8.54(s,1H),8.00(s,1H),7.62(d,J=8.7Hz,2H),7.30(d,J=8.2Hz,1H),7.27(s,1H),7.10(d,J=8.6Hz,2H),6.84(dd,J=8.3,2.0Hz,1H),6.76-6.73(m,1H),6.53(dd,J=7.9,1.9Hz,1H),4.11(t,J=6.3Hz,2H),3.73–3.69(m,4H),3.14–3.10(m,4H),2.70(t,J=7.3Hz,2H),2.07(dd,J=8.6,5.0Hz,2H).
Example 37
Synthesis of methyl 2- (4-oxo-6-phenoxy-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (37)
Figure BDA0002400750460000661
The title compound (37) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 35-c and 1-e in 80% yield.1H NMR(400MHz,DMSO-d6)δ9.70(s,1H),8.67(s,1H),8.32(s,1H),7.65–7.54(m,4H),7.51-7.45(m,2H),7.35–7.25(m,2H),7.19(d,J=7.7Hz,2H),4.76(d,J=17.1Hz,4H),4.33(s,2H),3.68(s,3H).
Example 38
Synthesis of methyl 2- (4-oxo-6-phenoxy-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (38)
Figure BDA0002400750460000662
The title compound (38) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 35-c and 16-a in 78% yield.1H NMR(400MHz,DMSO-d6)δ9.99(s,1H),8.62(s,1H),8.29(s,1H),7.63(d,J=8.7Hz,2H),7.50-7.44(m,2H),7.29–7.24(m,2H),7.16(d,J=7.8Hz,2H),7.10(d,J=8.6Hz,2H),4.77(s,2H),4.11(t,J=6.3Hz,2H),3.68(s,3H),2.72(t,J=7.2Hz,2H),2.11–2.02(m,2H).
Example 39
Synthesis of N- (3- (2- (4-fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-6-phenoxy-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (39)
Figure BDA0002400750460000671
The title compound (39) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 37 and 4-fluoropiperidine hydrochloride in 45% yield.1H NMR(400MHz,DMSO-d6)δ9.67(s,1H),8.66(s,1H),8.20(s,1H),7.56(d,J=9.8Hz,4H),7.50-7.44(m,2H),7.33(s,1H),7.31-7.26(m,1H),7.18(d,J=7.7Hz,2H),4.98–4.84(m,3H),4.74(s,2H),4.32(s,2H),3.67–3.59(m,1H),3.54–3.42(m,3H),2.04–1.91(m,1H),1.78(dd,J=10.0,6.9Hz,2H),1.69–1.58(m,1H).
Example 40
Synthesis of N- (3- (2- (3, 3-difluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-6-phenoxy-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (40)
Figure BDA0002400750460000672
The title compound (40) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 37 and 3, 3-difluoropiperidine hydrochloride in 55% yield.1H NMR(400MHz,DMSO-d6)δ9.68(s,1H),8.66(s,1H),8.21(d,J=4.0Hz,1H),7.56(d,J=9.1Hz,4H),7.50-7.44(m,2H),7.33(d,J=2.7Hz,1H),7.31-7.27(m,1H),7.18(d,J=7.8Hz,2H),4.92(d,J=20.8Hz,2H),4.74(s,2H),4.32(s,2H),3.91(t,J=11.7Hz,1H),3.78(t,J=12.1Hz,1H),3.56(d,J=4.7Hz,1H),3.52–3.44(m,1H),2.14–2.04(m,2H),1.77(s,1H),1.65–1.52(m,1H).
EXAMPLE 41
Synthesis of methyl 2- (6- (3-morpholinophenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (41)
Figure BDA0002400750460000681
The title compound (41) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 30-c and 16-a in 60% yield.1H NMR(400MHz,DMSO-d6)δ9.92(s,1H),8.59(s,1H),8.27(s,1H),7.63(d,J=8.7Hz,2H),7.31(d,J=8.2Hz,1H),7.27(s,1H),7.10(d,J=8.7Hz,2H),6.85(dd,J=8.4,2.0Hz,1H),6.76(s,1H),6.54(dd,J=8.0,1.8Hz,1H),4.77(s,2H),4.12(t,J=6.3Hz,2H),3.74–3.69(m,4H),3.68(s,3H),3.15–3.10(m,4H),2.72(t,J=7.2Hz,2H),2.07(p,J=6.8Hz,2H).
Example 42
Synthesis of N- (3- (2- (3, 3-difluoropiperidin-1-yl) -2-oxoethyl) -6- (3-morpholinophenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (42)
Figure BDA0002400750460000682
The title compound (42) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 30 and 3, 3-difluoropiperidine hydrochloride in 88% yield.1H NMR(400MHz,DMSO-d6)δ9.66(s,1H),8.65(s,1H),8.20(d,J=2.6Hz,1H),7.57(s,4H),7.34(d,J=2.8Hz,1H),7.31(d,J=8.2Hz,1H),6.87(dd,J=8.4,2.1Hz,1H),6.81(d,J=2.0Hz,1H),6.56(dd,J=8.0,2.0Hz,1H),4.94(s,1H),4.89(s,1H),4.75(s,2H),4.33(s,2H),3.91(t,J=11.5Hz,1H),3.76(d,J=12.2Hz,1H),3.71–3.68(m,4H),3.59–3.55(m,1H),3.52–3.47(m,1H),3.13–3.10(m,4H),2.09(dd,J=13.0,6.7Hz,2H),1.77(s,1H),1.61(s,1H).
Example 43
Synthesis of methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) -1, 4-dihydroquinazolin-3 (2H) -yl) acetate (43)
Figure BDA0002400750460000691
Methyl 2- (7-amino-6- (3- (dimethylamino) phenoxy) -4-oxa-1, 4-dihydroquinazolin-3 (2H) -yl) acetate (43-a)
Figure BDA0002400750460000692
Compound 10-b (1mmol) is dissolved in DCM and 37% aqueous formaldehyde (1mmol,1eq) and NaBH are added3CN (5mmol,5eq) and catalytic amount of acetic acid, at room temperature overnight. DCM/MeOH system column chromatography gave 43-a.
The title compound (43) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 43-a and 1-e in 85% yield.1H NMR(400MHz,DMSO-d6)δ9.20(s,1H),7.88(s,1H),7.56(d,J=7.9Hz,2H),7.44(d,J=8.0Hz,2H),7.17(s,1H),7.14-7.09(m,1H),6.93(s,1H),6.49(dd,J=8.3,2.1Hz,1H),6.38-6.35(m,1H),6.11(dd,J=7.9,2.0Hz,1H),4.67–4.64(m,4H),4.20(d,J=2.8Hz,4H),3.64(s,3H),2.84(s,6H).
Example 44
Synthesis of N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclohexane-1, 2' -quinazolin-7 ' -yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (44)
Figure BDA0002400750460000701
2, 4-diamino-5- (3- (dimethylamino) benzyl) benzamide (44-a)
Figure BDA0002400750460000702
Compound 25-a (1.52g) was dissolved in 8mL THF/8mL MeOH, Pd/C (1.52g) was added, and H was replaced three times2Then reacting at room temperature overnight to obtain the compound 44-a.
7 '-amino-6' - (3- (dimethylamino) phenoxy) -1 'H-spiro [ cyclohexane-1, 2' -quinazolin-4 '(3' H) -one (44-b)
Figure BDA0002400750460000703
Dissolving the compound 44-a (1mmol) in 10mL of acetic acid, adding cyclohexanone (1mmol,1eq) to react at 60 ℃ overnight, spin-drying, and performing DCM/MeOH column chromatography to obtain a compound 44-b.
The title compound (44) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 44-b and 1-e in 87% yield.1H NMR(400MHz,DMSO-d6)δ9.14(s,1H),7.96(s,1H),7.90(s,1H),7.56(d,J=8.1Hz,2H),7.43(d,J=8.0Hz,2H),7.12(dd,J=11.0,5.4Hz,2H),6.82(s,1H),6.48(dd,J=8.3,2.2Hz,1H),6.36-6.08(m,1H),6.10(dd,J=8.0,2.0Hz,1H),4.65(s,2H),4.19(s,2H),2.84(s,6H),1.78–1.71(m,2H),1.65-1.50(m,6H),1.48–1.35(m,2H).
Example 45
Synthesis of N- (7- (3- (dimethylamino) phenoxy) -3 a-methyl-1, 5-dioxo-1, 2,3,3a,4, 5-hexahydropyrrolo [1,2-a ] quinazolin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (45)
Figure BDA0002400750460000711
8-amino-7- (3- (dimethylamino) phenoxy) -3 a-methyl-2, 3,3a, 4-tetrahydropyrrole [1,2-a ] quinazoline-1, 5-dione (45-a)
Figure BDA0002400750460000712
According to the synthesis of compound 44-b described in example 44, 44-a was reacted with methyl levulinate to give 45-a.
The title compound (45) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 45-a and 1-e in 89% yield.1H NMR(400MHz,DMSO-d6)δ9.48(s,1H),9.08(s,1H),8.89(s,1H),7.60-7.49(m,4H),7.29(s,1H),7.25-7.15(m,1H),6.58(d,J=7.4Hz,1H),6.46(s,1H),6.28(d,J=7.3Hz,1H),4.71(s,2H),4.31-4.21(m,2H),2.87(s,6H),2.25-2.16(m,2H),1.43(s,3H),1.26-1.18(m,2H).
Example 46
Synthesis of N- (7- (3- (dimethylamino) phenoxy) -3 a-methyl-5-oxo-1, 2,3,3a,4, 5-hexahydropyrrolo [1,2-a ] quinazolin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (46)
Figure BDA0002400750460000721
8-amino-7- (3- (dimethylamino) phenoxy) -3 a-methyl-2, 3,3a, 4-tetrahydropyrrolo [1,2-a ] quinazolin-5 (1H) -one (46-a)
Figure BDA0002400750460000722
44-a and 5-chloro-2-pentanone were reacted according to the synthesis of compound 44-b described in example 44 to give 46-a.
The title compound (46) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 46-a and 1-e in 89% yield.1H NMR(400MHz,DMSO-d6)δ9.21(s,1H),8.30(s,1H),7.67(s,1H),7.56(d,J=8.0Hz,2H),7.44(d,J=7.9Hz,2H),7.22(s,1H),7.14-7.05(m,1H),6.49(dd,J=8.3,1.5Hz,1H),6.36(s,1H),6.14–6.09(m,1H),4.65(s,2H),4.20(s,2H),3.41(dd,J=17.6,14.0Hz,2H),2.84(s,6H),2.10-1.95(m,2H),1.26(s,3H),1.24-1.19(m,2H).
Example 47
Synthesis of 2- ((4-cyanobenzyl) oxa) -N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclopentane-1, 2' -quinazolin-7 ' -yl) acetamide (47)
Figure BDA0002400750460000731
7 '-amino-6' - (3- (dimethylamino) phenoxy) -1 'H-spiro [ cyclopentyl-1, 2' -quinazolin ] -4'(3' H) -one (47-a)
Figure BDA0002400750460000732
44-a was reacted with cyclopentanone to give 47-a according to the synthesis of compound 44-b described in example 44.
The title compound (47) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 47-a and 4-a in 80% yield.1H NMR(400MHz,DMSO-d6)δ9.12(s,1H),8.10(s,1H),7.85(s,1H),7.67(d,J=7.8Hz,2H),7.41(d,J=7.9Hz,2H),7.16–7.10(m,2H),6.93(s,1H),6.48(d,J=7.4Hz,1H),6.30(s,1H),6.10(d,J=7.6Hz,1H),4.63(s,2H),4.19(s,2H),2.85(s,6H),1.82-1.74(m,4H),1.69-1.61(m,4H).
Example 48
Synthesis of N- (6- (3- (dimethylamino) phenoxy) -2, 2-dimethyl-4-oxo-1, 2,3, 4-tetrahydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (48)
Figure BDA0002400750460000733
7-amino-6- (3- (dimethylamino) phenoxy) -2, 2-dimethyl-2, 3-dihydroquinazolin-4 (1H) -one (48-a)
Figure BDA0002400750460000741
44-a was reacted with acetone to give 48-a according to the synthesis of compound 44-b described in example 44.
The title compound (48) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 48-a and 1-e in 78% yield.1H NMR(400MHz,DMSO-d6)δ9.15(s,1H),7.96(s,1H),7.81(s,1H),7.56(d,J=7.8Hz,2H),7.43(d,J=7.8Hz,2H),7.17–7.06(m,2H),6.83(s,1H),6.48(d,J=8.0Hz,1H),6.35(s,1H),6.11(d,J=7.8Hz,1H),4.64(s,2H),4.19(s,2H),2.84(s,6H),1.37(s,6H).
Example 49
Synthesis of N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclopentane-1, 2' -quinazolin-7 ' -yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (49)
Figure BDA0002400750460000742
The title compound (49) was prepared according to the procedure for the synthesis of compound 1 described in example 1, using 47-a and 1-e in 85% yield.1H NMR(400MHz,DMSO-d6)δ9.15(s,1H),8.10(s,1H),7.87(s,1H),7.56(d,J=8.1Hz,2H),7.43(d,J=8.0Hz,2H),7.16–7.08(m,2H),6.93(s,1H),6.48(dd,J=8.4,2.1Hz,1H),6.36-6.33(m,1H),6.11(dd,J=8.0,1.9Hz,1H),4.65(s,2H),4.19(s,2H),2.84(s,6H),1.79-1.76(m,4H),1.70–1.62(m,4H).
Example 50
Synthesis of 4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline e-7-amide (50)
Figure BDA0002400750460000751
2-amino-5-iodoterephthalic acid dimethyl ester (50-a)
Figure BDA0002400750460000752
Dimethyl 2-aminoterephthalate (209mg,1mmol) and NIS (248mg, 1.1mmol) were dissolved in 20mL of acetic acid, stirred at room temperature for 24h, and after the conversion was complete the reaction was added to 20mL of ethyl acetate and 30mL of water. Adding saturated NaHCO3Adjusting the solution to neutral, washing the organic phase with water for 3 times, and MgSO4Drying and carrying out column chromatography to obtain 50-a.
4-amino-3 '- (trifluoromethyl) - [1,1' -diphenyl ] -2, 5-dicarboxylic acid (50-b)
Figure BDA0002400750460000753
Compound 50-a (1.68g) was mixed with 3-trifluoromethylphenylboronic acid (2.86g, 3eq), and Pd (PPh) was added3)4(0.58g, 10 mol%) and Ba (OH)3(7.03g, 5eq) dissolved in 12mL dioxane and 12mLH2And O, reacting at 80 ℃ overnight, adding EtOAC for dilution when the raw material disappears, adjusting the pH value to be acidic by using 6N hydrochloric acid, and performing column chromatography to obtain 50-b.
4-oxa-6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline-7-carboxylic acid (50-c)
Figure BDA0002400750460000754
The compound 50-b (2g) is dissolved in 10mL formamide, reacted at 160 ℃ overnight, cooled and subjected to suspension distillation column chromatography to obtain 50-c.
2- ((4- (trifluoromethyl) benzyl) oxy) ethylamine (50-d)
Figure BDA0002400750460000761
Dissolving p-trifluoromethyl benzyl bromide in dry DCM, adding N-boc-ethanolamine (2.5g, 0.67eq) and KOH (1.3g, 1eq) at 0 deg.C, returning to room temperature for reaction overnight, filtering, spin-drying, performing column chromatography, distilling under reduced pressure, adding 20mL CF3COOH and 40mLDCM react for 1h at room temperature, and then the reaction is finished, and the reaction is dried in a spinning mode to obtain 50-d.
The title compound 50 Compound 50-c (33.4mg) was dissolved in THF and 50-d (43.8mg, 2eq) was added) And HATU (114mg, 3eq) at room temperature overnight, spin-dried after completion of the reaction, and column chromatographed with 90% yield.1H NMR(400MHz,DMSO-d6)δ12.50(br s,1H),8.82-8.77(m,1H),8.21(s,1H),8.09(s,1H),7.73(dd,J=17.1,9.1Hz,6H),7.63-7.58(m,1H),7.54(d,J=8.0Hz,2H),4.53(s,2H),3.43(t,J=5.6Hz,2H),3.38–3.32(m,2H).
Example 51
Synthesis of methyl 2- (6- (3-chlorophenyl) -4-oxo-7- ((2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (51)
Figure BDA0002400750460000762
4-amino-3 '-chloro- [1,1' -diphenyl ] -2, 5-dicarboxylic acid (51-a)
Figure BDA0002400750460000763
According to the synthesis method of compound 50-b described in example 50, 50-a and 3-chlorobenzeneboronic acid were reacted to give 51-a.
Figure BDA0002400750460000771
51-b (87.3mg, 3eq) was dissolved in THF, triphosgene (29.7mg, 1eq) N was added2Reacting for 4h at 50 ℃ under protection until the raw materials disappear, dissolving the spin-dried solvent in water, adding glycine methyl ester hydrochloride (113mg, 9eq) and triethylamine (125 mu L, 9eq), reacting overnight, and obtaining 51-b after the complete spin-dried solvent is converted.
6- (3-chlorophenyl) -3- (2-methoxy-2-oxaethyl) -4-oxo-3, 4-dihydroquinazoline-7-carboxylic acid (51-c)
Figure BDA0002400750460000772
51-b (52mg) is dissolved in 10mL formic acid, reacted at 110 ℃ overnight, and subjected to spin-dry column chromatography after detecting no raw material to obtain 51-c.
The title compound (51) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 51-c and 50-d in 80% yield.1H NMR(400MHz,DMSO-d6)δ8.80-8.76(m,1H),8.47(s,1H),8.08(s,1H),7.72(s,2H),7.71(s,1H),7.56(d,J=8.0Hz,2H),7.49(s,1H),7.44–7.39(m,3H),4.89(s,2H),4.56(s,2H),3.72(s,3H),3.45(t,J=5.4Hz,2H),3.42–3.37(m,2H).
Example 52
Synthesis of 6- (3-chlorophenyl) -4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -3, 4-dihydroquinazoline e-7-amide (52)
Figure BDA0002400750460000773
6- (3-chlorophenyl) -4-oxo-3, 4-dihydroquinazoline-7-carboxylic acid (52-a)
Figure BDA0002400750460000781
51-a was reacted with formamide to give 52-a according to the synthesis of compound 50-c described in example 50.
The title compound (52) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 52-a and 50-d in 75% yield.1H NMR(400MHz,DMSO-d6)δ12.47(s,1H),8.78-8.74(m,1H),8.19(s,1H),8.05(s,1H),7.71(d,J=8.0Hz,2H),7.67(s,1H),7.55(d,J=8.0Hz,2H),7.47(d,J=1.5Hz,1H),7.40(q,J=4.4Hz,3H),4.56(s,2H),3.45(t,J=5.7Hz,2H),3.40–3.37(m,2H).
Example 53
Synthesis of N- (3- (4-chlorophenyl) propyl) -6- (3-fluorophenyl) -4-oxo-3, 4-dihydroquinazoline e-7-amide (53)
Figure BDA0002400750460000782
4-amino-3 '-fluoro- [1,1' -diphenyl ] -2, 5-dicarboxylic acid (53-a)
Figure BDA0002400750460000783
According to the synthesis method of the compound 50-b described in example 50, 50-a was reacted with 3-fluorobenzeneboronic acid to give 53-a.
6- (3-fluorophenyl) -4-oxo-3, 4-dihydroquinazoline-7-carboxylic acid (53-b)
Figure BDA0002400750460000791
43-a was reacted with formamide to give 53-b, following the synthesis of compound 50-c described in example 50.
The title compound (53) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 53-b and 4-chlorophenylpropylamine in 70% yield.1H NMR(400MHz,DMSO-d6)δ12.45(s,1H),8.59-8.54(m,1H),8.19(s,1H),8.06(s,1H),7.67(s,1H),7.45(dd,J=11.0,4.8Hz,1H),7.33(s,1H),7.31(d,J=3.4Hz,2H),7.29(d,J=1.6Hz,1H),7.21–7.18(m,2H),7.17(s,1H),3.15–3.10(m,2H),2.48–2.43(m,2H),1.65–1.59(m,2H).
Example 54
Synthesis of 6- (3-fluorophenyl) -4-oxo-N- (3-phenylpropyl) -3, 4-dihydroquinazoline e-7-amide (54)
Figure BDA0002400750460000792
The title compound (54) was prepared in 80% yield according to the procedure for the synthesis of compound 50 described in example 50, using 53-b and amphetamine.1H NMR(400MHz,DMSO-d6)δ12.45(s,1H),8.60-8.56(m,1H),8.19(s,1H),8.07(s,1H),7.67(s,1H),7.45(dd,J=14.3,8.0Hz,1H),7.32–7.29(m,2H),7.28–7.25(m,2H),7.23–7.19(m,1H),7.16(dd,J=5.9,4.7Hz,3H),3.16–3.10(m,2H),2.46(d,J=7.8Hz,2H),1.64(dd,J=14.6,7.3Hz,2H).
Example 55
Synthesis of 6- (3-chlorophenyl) -4-oxo-N- (3-phenylpropyl) -3, 4-dihydroquinazoline e-7-amide (55)
Figure BDA0002400750460000801
The title compound (55) was prepared in 78% yield according to the procedure for the synthesis of compound 50 described in example 50, using 52-a and amphetamine.1H NMR(400MHz,DMSO-d6)δ12.45(s,1H),8.62-8.58(m,1H),8.19(s,1H),8.06(s,1H),7.67(s,1H),7.49(s,1H),7.43(q,J=4.3Hz,3H),7.30-7.25(m,2H),7.19–7.14(m,3H),3.14(d,J=6.1Hz,2H),2.46(d,J=7.8Hz,2H),1.69–1.63(m,2H).
Example 56
Synthesis of 6- (3-chlorophenyl) -N- (3- (4-chlorophenyl) propyl) -4-oxo-3, 4-dihydroquinazoline-7-amide (56)
Figure BDA0002400750460000802
The title compound (56) was prepared in 80% yield according to the procedure for the synthesis of compound 50 described in example 50, using 52-a and 4-chlorophenylpropylamine.1H NMR(400MHz,DMSO-d6)δ12.45(s,1H),8.61-8.56(m,1H),8.19(s,1H),8.06(s,1H),7.67(s,1H),7.49(s,1H),7.45–7.41(m,3H),7.33(d,J=8.3Hz,2H),7.18(d,J=8.3Hz,2H),3.12(q,J=6.7Hz,2H),2.47–2.43(m,2H),1.63(dd,J=14.6,7.3Hz,2H).
Example 57
Synthesis of 4-oxo-N- (3-phenylpropyl) -6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline-7-amide (57)
Figure BDA0002400750460000803
The title compound (57) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 50-c and amphetamine in 85% yield.1H NMR(400MHz,DMSO-d6)δ12.50(s,1H),8.67-8.62(m,1H),8.21(s,1H),8.10(s,1H),7.77(d,J=11.3Hz,2H),7.71(d,J=8.2Hz,2H),7.66(t,J=7.5Hz,1H),7.26(t,J=7.3Hz,2H),7.17(d,J=7.1Hz,1H),7.13(d,J=7.6Hz,2H),3.11(dd,J=12.4,6.3Hz,2H),2.44(t,J=7.6Hz,2H),1.67–1.57(m,2H).
Example 58
Synthesis of N- (3- (4-chlorophenyl) propyl) -4-oxo-6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline-7-amide (58)
Figure BDA0002400750460000811
The title compound (58) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 50-c and 4-chlorophenylpropylamine in 86% yield.1H NMR(400MHz,DMSO-d6)δ12.50(s,1H),8.67-8.62(m,1H),8.21(d,J=2.2Hz,1H),8.10(s,1H),7.81–7.74(m,3H),7.72(d,J=7.4Hz,2H),7.69-7.64(m,1H),7.32(d,J=8.2Hz,2H),7.15(d,J=8.0Hz,2H),3.10(dd,J=12.3,6.2Hz,2H),2.42(t,J=7.6Hz,2H),1.59(dd,J=14.2,7.0Hz,2H).
Example 59
Synthesis of 6- (3-fluorophenyl) -4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -3, 4-dihydroquinazoline e-7-amide (59)
Figure BDA0002400750460000812
The title compound (59) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 53-b and 50-d in 80% yield.1H NMR(400MHz,DMSO-d6)δ12.46(s,1H),8.78-8.74(m,1H),8.19(s,1H),8.06(s,1H),7.71(d,J=7.9Hz,2H),7.67(s,1H),7.55(d,J=7.9Hz,2H),7.40(dd,J=14.5,7.4Hz,1H),7.30–7.25(m,2H),7.21-7.15(m,1H),4.55(s,2H),3.45(d,J=5.1Hz,2H),3.38(d,J=5.2Hz,2H).
Example 60
Synthesis of methyl 2- (6- (3-chlorophenyl) -7- ((3- (4-chlorophenyl) propyl) carbamoyl) -4-oxoquinazolin-3 (4H) -yl) acetate (60)
Figure BDA0002400750460000821
The title compound (60) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 51-c and 4-chlorophenylpropylamine in 83% yield.1H NMR(400MHz,DMSO-d6)δ8.62-8.58(m,1H),8.47(s,1H),8.08(s,1H),7.73(s,1H),7.50(s,1H),7.46–7.41(m,3H),7.33(d,J=8.3Hz,2H),7.18(d,J=8.3Hz,2H),4.89(s,2H),3.72(s,3H),3.13(dd,J=12.6,6.5Hz,2H),2.45(t,J=7.6Hz,2H),1.68–1.60(m,2H).
Example 61
Synthesis of methyl 2- (6- (3-fluorophenyl) -4-oxo-7- ((2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (61)
Figure BDA0002400750460000822
4-amino-3 '-fluoro-5- ((2-methoxy-2-oxoethyl) carbamoyl) - [1,1' -diphenyl ] -2-carboxylic acid (61-a)
Figure BDA0002400750460000831
53-a was reacted with glycine according to the synthesis of compound 51-b described in example 51 to give 61-a.
6- (3-fluorophenyl) -3- (2-methoxy-2-oxoethyl) -4-oxo-3, 4-dihydroquinazoline-7-carboxylic acid (61-b)
Figure BDA0002400750460000832
61-a was reacted with formic acid to give 61-b according to the synthesis of compound 51-c described in example 51.
The title compound (61) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 61-b and 50-d in 89% yield.1H NMR(400MHz,DMSO-d6)δ8.80-8.75(m,1H),8.47(s,1H),8.08(s,1H),7.73(s,1H),7.71(s,2H),7.56(d,J=7.7Hz,2H),7.41(d,J=6.7Hz,1H),7.29(d,J=7.9Hz,2H),7.22-7.17(m,1H),4.89(s,2H),4.56(s,2H),3.72(s,3H),3.49–3.40(m,4H).
Example 62
Synthesis of methyl 2- (6- (3-fluorophenyl) -4-oxo-7- ((3-phenylpropyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (62)
Figure BDA0002400750460000833
The title compound (62) was prepared in 92% yield according to the procedure for the synthesis of compound 50 described in example 50, using 61-b and amphetamine.1H NMR(400MHz,DMSO-d6)δ8.61(s,1H),8.47(s,1H),8.09(s,1H),7.72(s,1H),7.46(dd,J=14.5,7.6Hz,1H),7.29(dd,J=14.9,7.3Hz,4H),7.18(dd,J=18.7,8.0Hz,4H),4.89(s,2H),3.72(s,3H),3.14(dd,J=12.4,6.3Hz,2H),2.46(d,J=7.8Hz,2H),1.70–1.60(m,2H).
Example 63
Synthesis of methyl 2- (7- ((3- (4-chlorophenyl) propyl) carbamoyl) -6- (3-fluorophenyl) -4-oxoquinazolin-3 (4H) -yl) acetate (63)
Figure BDA0002400750460000841
The title compound (63) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 61-b and 4-chlorophenylpropylamine in 94% yield.1H NMR(400MHz,DMSO-d6)δ8.60(d,J=4.8Hz,1H),8.47(s,1H),8.09(s,1H),7.73(s,1H),7.46(dd,J=14.5,7.7Hz,1H),7.31(dd,J=16.3,7.9Hz,4H),7.22(d,J=8.7Hz,1H),7.18(d,J=8.1Hz,2H),4.89(s,2H),3.72(s,3H),3.13(dd,J=12.0,6.0Hz,2H),2.45(t,J=7.5Hz,2H),1.63(dd,J=13.9,6.9Hz,2H).
Example 64
Synthesis of methyl 2- (6- (3-chlorophenyl) -4-oxo-7- ((3- (4- (trifluoromethyl) phenoxy) propyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (64)
Figure BDA0002400750460000842
3- (4- (trifluoromethyl) phenoxy) propyl-1-amine (64-a)
Figure BDA0002400750460000843
P-trifluoromethylphenol and N-boc-propanolamine were reacted according to the method for synthesizing compound 50-d described in example 50 to give 64-a.
The title compound (64) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 51-c and 64-a in 88% yield.1H NMR(400MHz,DMSO-d6)δ8.70-8.65(m,1H),8.46(s,1H),8.08(s,1H),7.74(s,1H),7.65(d,J=8.7Hz,2H),7.50(s,1H),7.47–7.38(m,3H),7.05(d,J=8.6Hz,2H),4.89(s,2H),3.89(t,J=6.1Hz,2H),3.72(s,3H),3.33–3.28(m,2H),1.87–1.79(m,2H).
Example 65
Synthesis of 6- (3-chlorophenyl) -4-oxo-N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (65)
Figure BDA0002400750460000851
The title compound (65) was prepared according to the procedure for the synthesis of compound 50 described in example 50, using 52-a and 64-a in 86% yield.1H NMR(400MHz,DMSO-d6)δ8.66-8.63(m,1H),8.19(s,1H),8.06(s,1H),7.69(s,1H),7.64(d,J=8.4Hz,2H),7.48(s,1H),7.42(dd,J=6.0,3.3Hz,2H),7.39(dd,J=5.6,2.7Hz,1H),7.36–7.23(m,1H),7.04(d,J=8.8Hz,2H),3.89(t,J=6.1Hz,2H),3.27(dd,J=12.6,6.2Hz,2H),1.87–1.79(m,2H).
Example 66
Synthesis of 2- (6- (3-chlorophenyl) -4-oxo-7- ((3- (4- (trifluoromethyl) phenoxy) propyl) carbamoyl) quinazolin-3 (4H) -yl) acetic acid (66)
Figure BDA0002400750460000861
The title compound (66) Compound 64(1eq) was dissolved in 6mL THF and 3mLH2And adding LiOH (3eq) into the mixture to react at room temperature overnight, performing spin drying after the reaction is finished, adding 1N hydrochloric acid to adjust the pH value to be weakly acidic, separating out a large amount of white solid, filtering and drying, and obtaining the yield of 85%.1H NMR(400MHz,DMSO-d6)δ8.67-8.64(m,1H),8.45(s,1H),8.08(s,1H),7.73(s,1H),7.65(d,J=8.7Hz,2H),7.50(s,1H),7.43(dd,J=5.9,3.0Hz,2H),7.40(dd,J=4.6,1.9Hz,1H),7.05(d,J=8.7Hz,2H),4.76(s,2H),3.89(t,J=6.1Hz,2H),3.29–3.20(m,2H),1.83(dd,J=12.6,6.3Hz,2H).
Example 67
Synthesis of 6- (3-chlorophenyl) -4-oxo-3- (2-oxo-2- (piperidin-1-yl) ethyl) -N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (67)
Figure BDA0002400750460000862
The title compound (67) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 66 and cyclohexylamine, yield 86%.1H NMR(400MHz,DMSO-d6)δ8.68-8.64(m,1H),8.40(d,J=49.8Hz,1H),8.07(d,J=8.1Hz,1H),7.73(d,J=8.7Hz,1H),7.65(d,J=8.7Hz,2H),7.51–7.48(m,1H),7.44–7.42(m,2H),7.41–7.36(m,1H),7.05(d,J=8.7Hz,2H),4.92(d,J=27.1Hz,2H),3.89(t,J=6.1Hz,2H),3.72(s,2H),3.46(dd,J=18.5,13.5Hz,4H),3.31(q,J=6.3Hz,2H),1.87–1.81(m,2H),1.54(d,J=62.5Hz,4H).
Example 68
Synthesis of 6- (3-chlorophenyl) -3- (2- (3, 3-difluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (68)
Figure BDA0002400750460000871
The title compound (67) was prepared in 80% yield according to the procedure for the synthesis of compound 20 described in example 20, using 66 and 3, 3-difluorocyclohexylamine.1H NMR(400MHz,DMSO-d6)δ8.68-8.64(m,1H),8.35(d,J=5.4Hz,1H),8.06(s,1H),7.73(s,1H),7.65(d,J=8.7Hz,2H),7.49(s,1H),7.45(dd,J=9.6,5.1Hz,1H),7.42–7.38(m,2H),7.05(d,J=8.6Hz,2H),5.01(d,J=21.1Hz,2H),3.99–3.77(m,4H),3.63–3.58(m,1H),3.53(dd,J=6.9,2.7Hz,1H),3.32–3.24(m,2H),2.20–2.03(m,2H),1.83(dd,J=11.8,5.5Hz,3H),1.67–1.56(m,1H).
Example 69
Synthesis of N- (6- (4- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (69)
Figure BDA0002400750460000872
The title compound (69) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 33 and propargylamine in 75% yield.
Example 70
Synthesis of N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (70)
Figure BDA0002400750460000881
The title compound (70) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 10 and propargylamine in 70% yield.1H NMR(400MHz,DMSO-d6)δ9.66(s,1H),8.78-8.74(m,1H),8.64(s,1H),8.24(s,1H),7.57(s,3H),7.34(s,1H),7.27-7.22(m,1H),6.63(dd,J=8.2,2.4Hz,1H),6.53-6.51(m,1H),6.36(dd,J=7.7,2.1Hz,1H),4.75(s,2H),4.60(s,2H),4.33(s,2H),3.89(dd,J=5.4,2.5Hz,2H),3.31,(s,1H),3.18-3.15(m,1H),2.89(s,6H).
Example 71
Synthesis of N- (6- (3-morpholinophenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (71)
Figure BDA0002400750460000882
The title compound (71) was prepared according to the procedure for the synthesis of compound 20 described in example 20, using 30 and propargylamine, yield 78%.1H NMR(400MHz,DMSO-d6)δ9.66(s,1H),8.78(d,J=4.3Hz,1H),8.65(d,J=4.1Hz,1H),8.25(d,J=4.1Hz,1H),7.70(dd,J=13.4,3.9Hz,1H),7.58(d,J=4.2Hz,3H),7.34–7.26(m,2H),6.89–6.84(m,1H),6.81(d,J=2.0Hz,1H),6.58–6.53(m,1H),4.75(s,2H),4.60(s,2H),4.32(s,2H),3.89(d,J=2.5Hz,2H),3.70(s,4H),3.20–3.15(m,1H),3.11(s,4H).
[ pharmacological experiments ]
The test methods used in the following examples are conventional methods unless otherwise specified.
Materials, reagents and the like used in the following examples are commercially available unless otherwise specified.
Experimental example 1 investigation of the antagonistic action of 1130-2 (Compound 68 prepared in example 68) on NOD1 and NOD2
Materials and methods
(1) Cells and reagents
The test substance: compounds were formulated as 20mM DMSO stocks and diluted with medium to the appropriate concentrations just prior to use.
Cell: HEK-Blue hNOD1 cell, HEK-Blue hNOD2 cell, and 293-hNOD2 were purchased from InvivoGen.
Antibody: beta-actin (Abcam, ab6276), I.kappa.B alpha (CST, 9242S), JNK (CST, 9252S), p-JNK (CST, 9251S), p38(CST, 9212S), p-p38(CST, 4631S), ERK (Santa cruz, sc-94), p-ERK (Santa cruz, sc-7383), RIP2(CST, 4142), p-RIP2(CST, 4364), Goat polymeric binder Antibody to Mouse-H & L-HRP (Abcam, ab6721), polymeric binder Antibody to Mouse-H & L-HRP (Abcam, 6728)
Primers and probes: primers and probes for IL-6(Hs00985639_ m1), IL-8(Hs00174103_ m1) and GAPDH (Hs02758991_ g1) were purchased from Invitrogen
Reagent: the hNOD1 agonist C12-ie-DAP (the addition of a lauryl group to the glutamic acid residue of ie-DAP), the hNOD2 agonist Muramyl Dipeptide (MDP), HEK-Blue Detection, Normocin, Blastidin and Zeocin are all available from InvivoGen; DMEM medium, RPMI1640 medium, fetal bovine serum, Penicilin-Streptomyces, PBS, EDTA, Trizol, High-capacity cDNA Reverse Transcription Kits,
Figure BDA0002400750460000891
gene Expression Master Mix was purchased from Invitrogen; RNeasy Mini Kit was purchased from QIAGEN; phosphatase inhibitor Cocktail, protease inhibitor, Na3VO4, SRB, TCA, Tris-base and glacial acetic acid were purchased from Sigma; NaF and BSA were purchased from Amresco; skim Milk from BD; clarityTMWestern ECL substrate, PVDF membrane and Precision Plus Protein Dual color standards from Bio-Rad; 3MM filter paper from Millipore; RIPA lysate, SDS-PAGE Gel Kit, Loading buffer, membrane buffer, running buffer and regeneration solution of Western blotting membrane were purchased from Yukang as a century.
(2) Instrument and consumable
Microplate reader (POLARstar Omega, USA), horizontal centrifuge (Eppendorf), microscope (OLYMPUS), CO2Cell incubator (Thermo fisher), Nanodrop spectrophotometry (Thermo fisher), fluorescent quantitative PCR instrument (ABI), PCR instrument (Eppendorf), electrophoresis instrument (Bio-Rad), transfer tank (Bio-Rad), ChemiDocTM XRS+Imager (Bio-Rad), blood count plate (Shanghai Biochemical instruments Co., Ltd.), shaker, etc. 96-well plates (Costar, USA), 6-well plates (Costar, USA), 12-well plates (Costar, USA), centrifuge tubes (Corning), petri dishes (Corning), and the like.
(3) Detection of HEK-Blue hNOD1 antagonist (HEK-Blue Detection)
HEK-Blue hNOD1 cells were cultured in DMEM medium (DMEM complete medium) containing 10% FBS, 50U/mL penicillin, 50. mu.g/mL streptomycin, 50. mu.g/mL Normolin, 2mM L-glutamine. Collecting cells in logarithmic growth phase, HEK-Blue Detection testThe assay solution was adjusted for cell concentration and inoculated into 96-well plates (50000/well) and a gradient working solution of test compound (final concentrations 10. mu.M, 5. mu.M, 2.5. mu.M, 1.25. mu.M, 625nM, 312.5nM, 31.25nM, 3.125nM, 0.3125nM and vehicle control, 37 ℃ C., 5% CO) was added2After 3h incubation in the incubator, the positive stimulus C12-ie-DAP (final concentration 50ng/mL), 37 ℃, 5% CO was added2The incubation was continued in the incubator for 20h and the OD was measured at 655 nm.
The positive control group OD value was designated as C, and the test group OD value was designated as (T).
Antagonism Rate% (Inhibit Rate) [ (C-T)/C]X 100, calculating IC50I.e., [ (C-T)/C]Compound concentration at x 100-50;
and after the detection is finished, the influence of the compound on the cell growth is inspected by adopting an SRB method. 50 mu L/well is added with 80 percent of TCA and fixed for 1h at 4 ℃; washing the plate with deionized water for 5 times, and naturally drying; adding 0.4% SRB into 100 μ L/well for dyeing, and standing at room temperature for 10 min; washing the plate with 100 μ L/well 1% acetic acid for 5 times, and naturally drying; adding Tris base into the well at a rate of 150. mu.L/well, and oscillating for about 5 min; detecting an OD value at 515nm of an enzyme-labeling instrument; the OD value of the vehicle control group is denoted as C, and the OD value of the test group is denoted as (T). Growth rate% (percent Growth) [ T/C ] × 100.
(4) Detection of HEK-Blue hNOD2 antagonist (HEK-Blue Detection)
The method is the same as the detection of an antagonist of HEK-Blue hNOD 1. The cell density was 25000 cells/well, the positive stimulus was MDP (50ng/mL), the incubation time was 18h, and the detection wavelength was 650 nm.
(5) Reverse transcription and fluorescent quantitative PCR
Collecting THP1 cells in logarithmic growth phase, adjusting cell concentration to 1.25 × 106Per well density was inoculated into 6-well plates, compound 1130-2 (final concentrations of 10. mu.M and 1. mu.M, respectively) or a positive control (positive control, 1. mu.M) was added, after 1 hour of incubation, C12-ie-DAP (final concentration of 500ng/mL) was added for 90min, and after lysis of the cell sample with Trizol, total RNA was extracted and purified with RNAeasy mini kit (Qianen). The RNA content is detected by a Nanodrop spectrophotometer (Thermo), the ratio of the absorbance of each sample at a wavelength of 260 to the absorbance at a wavelength of 280 (A260:280OD rates) is more than 1.8,1000ng of RNA was reverse transcribed into cDNA. Adding corresponding primers and probes into 1 mu L of cDNA, adding the cDNA into a 96-well plate with a final volume of 20 mu L, and placing the cDNA in a fluorescent quantitative PCR instrument for reaction. IL-6, IL-8 and GAPDH primers and probes were purchased from Invitrogen. The reaction system is as follows: 10 μ L TaqMan Gene Expression Master Mix (2X), 1 μ L TaqMan Gene Expression Assay (20X), 2 μ L cDNA, 7 μ L dH2O。
The reaction parameters are as follows:
①50℃,2min
②95℃,10min
③95℃,15sec
④60℃,1min③to④40cycles
GAPDH was used as reference gene, 2-△ctThe relative expression amount is calculated, and the antagonism rate is calculated.
(6)Western Blotting
Cells from log phase growth were harvested, plated in 6-well plates, and test compound 1130-2 (final concentrations of 10. mu.M and 1. mu.M), NOD1 positive antagonist (final concentration of 1. mu.M), 37 ℃, 5% CO were added2After incubation in an incubator for 1h, the stimulus C12-ie-DAP (final concentration of 500ng/mL) was added, and after incubation for 30min or 45min, the RIPA lysate was used to lyse the cells and extract the proteins, which were subjected to SDS-PAGE at a loading of 15. mu.L/well. After electrophoresis, proteins are transferred to a PVDF membrane, the PVDF membrane is sealed by 5% of skimmed milk powder at room temperature for 2 hours, and primary antibodies diluted in a proper proportion are respectively added for overnight at 4 ℃. Rinsing with 1 XTSST for 3 times, adding diluted secondary antibody, incubating at room temperature for 1h, rinsing with 1 XTSST, adding ECL luminescent substrate, ChemiDocTM XRS+Exposure in the imager.
Test results
As shown in FIG. 4, compound 1130-2 has significant antagonistic effect on hNOD1 and hNOD2 signaling pathways, IC501.127 and 0.7694 respectively. As can be seen from FIG. 5, 1130-2 can significantly antagonize the agonistic action of C12-ie-DAP on NOD1 and NOD2 signaling pathways of THP1 cells, and reduce the transcription of inflammatory cytokines IL-6, TNF alpha and IL-8 mRNA.
As can be seen in FIG. 6, THP1 cells stimulated with C12-ie-DAP or MDP showed increased expression of phosphorylated RIP2, increased expression of phosphorylated IKK α/β, P65, and decreased expression of IkappaB α, indicating activation of NF-kappaB signaling pathway, as compared to vehicle Control (Control); increased expression of phosphorylated JNK, p38, indicates that the MAPK signaling pathway is activated. After the cells are treated by 1130-2, compared with a C12-ie-DAP single group, the expression of phosphorylated RIP2 is reduced, the expression of phosphorylated IKK alpha/beta and P65 is reduced, the expression of IkB alpha is increased, and the expression of phosphorylated JNK and P38 is reduced, so that 1130-2 can block the activation of NF-kappa B and MAPK signal channels mediated by NOD1 and NOD 2.
In conclusion, 1130-2 can significantly antagonize NOD1/2 mediated activation of the signaling pathway and is an NOD1/2 antagonist.
Experimental example 2
Antagonism of NOD1 and NOD2 in vitro by the Compounds prepared in examples 1-71
Table 1 details the percent antagonism of the NOD1/2 signaling pathway for the compounds of the examples. Wherein 10. mu.M represents the percent antagonism of the NOD1/2 pathway at a test compound concentration of 10. mu.M, and 5. mu.M represents the percent antagonism of the NOD1/2 pathway at a test compound concentration of 5. mu.M. IC (integrated circuit)50Values represent the concentration of test compound at a percent antagonism of 50%; "ND" means not detected.
TABLE 1 antagonism of NOD1 and NOD2 in vitro by the compounds of the examples
Figure BDA0002400750460000921
Figure BDA0002400750460000931
Figure BDA0002400750460000941
Figure BDA0002400750460000951
Figure BDA0002400750460000961
Experimental example 3
Effect of 1130-2 (Compound 68 prepared in example 68) on growth of mouse B16-F10 melanoma model tumors
Test article
PTX, 1130-2 were prepared as 20 × DMSO stock solutions, which were diluted to dosing concentrations with 5% polyoxyethylene castor oil (Cremophor EL) and 5% DMSO, respectively, just before use.
Experimental animals and tumor strains:
male C57BL/6 mice, 6-8 weeks old, were purchased from Beijing Wittingle laboratory animal technology, Inc., license number SCXK (Jing) 2012-0001, and were bred at the animal center of Qinghua university. The tumor cells used in the assay were: mouse B16-F10 melanoma cells, purchased from ATCC.
The test method comprises the following steps:
B16-F10 melanoma cells were cultured in DMEM medium with 10% FBS, 50U/mL penicillin, 50. mu.g/mL streptomycin. Taking healthy C57BL/6 male mice axilla subcutaneous inoculation tumor liquid, 2X 105The inoculation volume is 0.1ml per cell. Assuming that the day of inoculation is D0, on day 1 after inoculation (D1), mice were randomly divided into 4 groups by weight, each: vehicle Control group (Control); 1130 and 250mg/kg group; ③ 12mg/kg group of Paclitaxel (PTX); 1130-2(50mg/kg) + PTX (12 mg/kg). The PTX 12mg/kg intermittent administration group animals are administrated by intravenous injection for 1 time every 4 days, and the administration time is 4 times; 1130-2 and vehicle control group animals are administered by tail vein injection for 1 time every 2 days, 8 times in total; 1130-2(50mg/kg) + PTX (12mg/kg) animals in the group were injected intravenously with PTX (12mg/kg)1 time every 4 days and 1130-2(50mg/kg)1 time every 2 days.
During the administration, the animal body weight is measured every 2 days, and the length and the short diameter of the tumor are measured by a vernier caliper according to the formula: (1/2) × major axis × (minor axis)2Tumor size was calculated. The experiment was terminated on day 16 after inoculation (D16), and the mice were sacrificed by cervical dislocation after blood sampling from the eyeballs, dissected, removed the tumor, and weighed. Comparative analysis of the administered group and the control group was performed.
And (3) test results:
as shown in FIG. 7, the tumor volume growth rate of the 1130-2(50mg/kg) single administration group was comparable to that of the vehicle control group, indicating that the 1130-2 single administration did not affect the tumor growth. PTX (12mg/kg) and 1130-2(50mg/kg) + PTX (12mg/kg) groups all can significantly inhibit the growth of tumors, and have statistical significance (p is less than 0.001), and the tumor weight inhibition rates of the two groups are 65.1% and 85.5%, respectively. The combined group of 1130-2(50mg/kg) + PTX (12mg/kg) has obviously stronger tumor inhibition effect than that of a PTX (12mg/kg) single-acting group, and has statistical significance (p is less than 0.01).
In the description herein, references to the description of the term "one embodiment," "some embodiments," "an example," "a specific example," or "some examples," etc., mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above are not necessarily intended to refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, various embodiments or examples and features of different embodiments or examples described in this specification can be combined and combined by one skilled in the art without contradiction.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that variations, modifications, substitutions and alterations can be made to the above embodiments by those of ordinary skill in the art within the scope of the present invention.

Claims (10)

1. The compound shown as a general formula (I) or (II), and a stereoisomer and a medicinal salt thereof,
Figure FDA0002400750450000011
wherein:
R1、R2independently selected from hydrogen, C1-6Alkyl radical, R1、R2Can form a quaternary, five-membered or six-membered aliphatic heterocyclic ring with nitrogen at position 1 and/or carbon atom at position 2 of the mother nucleus of the compound of the general formula II independently or together; the quaternary, quinary or hexahydric aliphatic heterocyclic ring can be substituted or unsubstituted, and the substituent on the quaternary, quinary or hexahydric aliphatic heterocyclic ring can be one or more and is independently selected from the following groups: o, halogen atom, hydroxyl group, mercapto group, amino group, aldehyde group, carboxyl group, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, carbamoyl, nitro, cyano;
R3selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3,CO(CH2)3CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-6Alkyl, and, R312And R313Can form a quaternary, quinary or hexahydric aliphatic heterocyclic ring with nitrogen atoms;
R6is selected from
Figure FDA0002400750450000021
Wherein:
indicates the position of the bond
R61Represents one or more substituents independently selected from: hydrogen, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl, C1-6 alkoxy substituted C1-6 alkyl, carbamoyl, nitro, cyano, four-, five-or six-membered aliphatic ring or aliphatic heterocycle;
R7is selected from
Figure FDA0002400750450000022
Wherein:
R71optionally substituted or unsubstituted five-membered, six-membered aromatic ring or five-membered, six-membered aromatic heterocycle; the substituent on the substituted five-membered, six-membered aromatic ring or five-membered, six-membered aromatic heterocyclic ring may be one or more, and is arbitrarily selected from: : hydrogen, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl, C1-6 alkoxy substituted C1-6 alkyl, carbamoyl, nitro, cyano.
g is an integer optionally selected from 0, 1,2,3 or 4, and represents CH when g is selected from 02Is absent;
h is an integer selected from 0, 1,2,3 or 4, and when h is selected from 0, it represents CH2Is absent;
y is optionally selected from carbon, nitrogen, oxygen, sulfur atoms or Y is absent.
2. A compound according to claim 1, and stereoisomers and pharmaceutically acceptable salts thereof, wherein said compound is of formula IA
Figure FDA0002400750450000023
Figure FDA0002400750450000031
Wherein:
R3selected from hydrogen or- (CH) mR32(CH2)nR31Which isThe method comprises the following steps:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3,CO(CH2)3CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-6Alkyl radical, C1-6Alkylene radical, C1-6Alkynyl, and, R312And R313Can form a quaternary, quinary or hexahydric aliphatic heterocyclic ring with nitrogen atoms;
R61represents one or more substituents independently selected from: hydrogen, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl radical, C1-6Alkoxy-substituted C1-6Alkyl, carbamoyl, nitro, cyano, quaternary, quinary or senary aliphatic or aliphatic heterocyclic ring;
g is an integer selected from 0, 1,2 or 3, and when g is selected from 0, it represents CH2Is absent;
h is an integer selected from 0, 1,2 or 3, and when h is selected from 0, it represents CH2Is absent;
y is optionally selected from oxygen, sulfur atoms or Y is absent;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
3. The compound of claim 1, and stereoisomers and pharmaceutically acceptable salts thereof, wherein said compound is represented by formula IB
Figure FDA0002400750450000041
R3Selected from hydrogen or- (CH) mR32(CH2)nR31Wherein:
m is selected from the group consisting of 0 or 1,
R32is selected from COOCH3,COOCH2CH3,CO(CH2)2CH3,CO(CH2)3CH3
When m is selected from 0, represents CH and R32Are not present at all and are,
n is an integer selected from 1 to 4,
R31independently selected from-COOR311or-CONR312R313
Wherein R is311、R312And R313Each independently selected from hydrogen atom, C1-6Alkyl radical, C1-6Alkylene radical, C1-6Alkynyl, and, R312And R313Can form a quaternary, quinary or hexahydric aliphatic heterocyclic ring with nitrogen atoms;
R61represents one or more substituents independently selected from: hydrogen, halogen atom, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, C1-6Alkyl radical, C1-6Alkoxy radical, C1-6Alkanemercapto group, C1-6Alkylamino, halogen-substituted C1-6Alkyl, C1-6 alkoxy substituted C1-6 alkyl, carbamoyl, nitro, cyano, four-, five-or six-membered aliphatic ring or aliphatic heterocycle;
g is an integer selected from 0, 1,2 or 3, and when g is selected from 0, it represents CH2Is absent;
h is an integer selected from 0, 1,2 or 3, and when h is selected from 0, it represents CH2Is absent;
y is optionally selected from oxygen, sulfur atoms or Y is absent;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-4Alkyl radical, C1-4Alkoxy radical, C1-4Alkanemercapto group, C1-4Alkylamino, halogen-substituted C1-4Alkyl, carbamoyl, nitro, cyano.
4. A compound according to claim 2, and stereoisomers and pharmaceutically acceptable salts thereof, wherein said compound is represented by formula IA4
Figure FDA0002400750450000051
R314Represents one or more substituents independently selected from: hydrogen, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano, six-membered aliphatic heterocycle;
R711represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano.
5. A compound according to claim 3, and stereoisomers and pharmaceutically acceptable salts thereof, wherein said compound is represented by formula IB2b
Figure FDA0002400750450000052
R314Represents one or more substituents independently selected from: hydrogen, oxygen atom, fluorine, chlorine, bromine, iodine, hydroxyl, sulfydryl, amino, aldehyde group, carboxyl and C1-3Alkyl radical, C1-3Alkoxy radical, C1-3Alkanemercapto group, C1-3Alkylamino, halogen-substituted C1-3Alkyl, carbamoyl, nitro, cyano;
R61represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde, carboxyl, methyl, ethyl, methoxyl, ethoxyl, methylmercapto, ethylsulfhydryl, methylamino, ethylamino, dimethylamino, diethylamino,
Figure FDA0002400750450000061
R711Represents one or more substituents independently selected from: hydrogen, fluorine, chlorine, bromine, iodine, hydroxyl, sulfhydryl, amino, aldehyde group, carboxyl, methyl, ethyl, propyl, methoxyl, ethoxyl, propoxy, methylmercapto, ethylsulfhydryl, propylsulfhydryl, methylamino, ethylamino, propylamino, CF3Carbamoyl, nitro, cyano.
6. The compound according to any one of claims 1-5, and stereoisomers and pharmaceutically acceptable salts thereof, wherein said compound is selected from the group consisting of:
dimethyl (R) -2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) glutarate (Compound 1)
Dimethyl (R) -2- (7- (2- ((2-chloro-4-fluorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 2)
5-Ethyl-1-methyl (R) -2- (7- (2- ((2-chloro-4-fluorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 3)
Dimethyl (R) -2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 4)
5-Ethyl-1-methyl (R) -2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 5)
Dimethyl (R) -2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 6)
5-Ethyl-1-methyl (R) -2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 7)
Dimethyl (R) -2- (7- (2- ((3, 4-dichlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 8)
5-Ethyl-1-methyl (R) -2- (7- (2- ((3, 4-dichlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) glutarate (Compound 9)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 10)
Methyl 2- (7- (2- (Benzyloxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 11)
Methyl 2- (7- (2- ((4-cyanobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 12)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -7- (2- ((4-fluorobenzyl) oxa) acetoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 13)
Methyl 2- (7- (2- ((4-chlorobenzyl) oxa) acetoxy) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 14)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -7- (4- (4-fluorophenoxy) butanamide) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 15)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (Compound 16)
Methyl 2- (7- (4- (4-cyanophenoxy) butanamide) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 17)
Methyl 2- (7- (4-cyanobenzoylamino) -6- (3- (dimethylamino) phenoxy) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 18)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (4- (trifluoromethyl) benzoylamino) quinazolin-3 (4H) -yl) acetate (Compound 19)
N- (3- (2- (3, 3-Difluoropiperidin-1-yl) -2-oxoethyl) -6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 20)
N- (3- (2- (4, 4-Difluoropiperidin-1-yl) -2-oxoethyl) -6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 21)
N- (6- (3- (dimethylamino) phenoxy) -3- (2- (4-fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 22)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (piperidin-1-yl) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 23)
N- (6- (3- (dimethylamino) phenoxy) -3- (2- (4-fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4-fluorophenoxy) butanamide (compound 24)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 25)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4- (trifluoromethyl) phenoxy) butanamide (Compound 26)
2- ((4-cyanobenzyl) oxa) -N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) acetamide (Compound 27)
3-cyano-N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) benzamide (compound 28)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (trifluoromethyl) benzamide (compound 29)
Methyl 2- (6- (3-Morpholphenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 30)
N- (6- (3-Morpholphenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 31)
Methyl 2- (6- (2- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 32)
Methyl 2- (6- (4- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 33)
Methyl 2- (6- (4- (dimethylamino) phenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (compound 34)
Methyl 2- (7- (2- ((2, 4-dichlorobenzyl) oxa) acetoxy) -4-oxo-6-phenoxyquinazolin-3 (4H) -yl) acetate (Compound 35)
N- (6- (3-Morpholinophenoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -4- (4- (trifluoromethyl) phenoxy) butanamide (Compound 36)
Methyl 2- (4-oxo-6-phenoxy-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) quinazolin-3 (4H) -yl) acetate (Compound 37)
Methyl 2- (4-oxo-6-phenoxy-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (Compound 38)
N- (3- (2- (4-Fluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-6-phenoxy-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 39)
N- (3- (2- (3, 3-Difluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-6-phenoxy-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 40)
Methyl 2- (6- (3-morpholinophenoxy) -4-oxo-7- (4- (4- (trifluoromethyl) phenoxy) butanamide) quinazolin-3 (4H) -yl) acetate (Compound 41)
N- (3- (2- (3, 3-Difluoropiperidin-1-yl) -2-oxoethyl) -6- (3-morpholineoxy) -4-oxo-3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 42)
Methyl 2- (6- (3- (dimethylamino) phenoxy) -4-oxo-7- (2- ((4- (trifluoromethyl) benzyl) oxa) acetoxy) -1, 4-dihydroquinazolin-3 (2H) -yl) acetate (Compound 43)
N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclohexane-1, 2' -quinazolin-7 ' -yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 44)
N- (7- (3- (dimethylamino) phenoxy) -3 a-methyl-1, 5-dioxo-1, 2,3,3a,4, 5-hexahydropyrrolo [1,2-a ] quinazolin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 45)
N- (7- (3- (dimethylamino) phenoxy) -3 a-methyl-5-oxo-1, 2,3,3a,4, 5-hexahydropyrrolo [1,2-a ] quinazolin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 46)
2- ((4-cyanobenzyl) oxa) -N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclopentane-1, 2' -quinazolin-7 ' -yl) acetamide (Compound 47)
N- (6- (3- (dimethylamino) phenoxy) -2, 2-dimethyl-4-oxo-1, 2,3, 4-tetrahydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 48)
N- (6' - (3- (dimethylamino) phenoxy) -4' -oxo-3 ',4' -dihydro-1 ' H-spiro [ cyclopentane-1, 2' -quinazolin-7 ' -yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 49)
4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline e-7-amide (Compound 50)
Methyl 2- (6- (3-chlorophenyl) -4-oxo-7- ((2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (Compound 51)
6- (3-chlorophenyl) -4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -3, 4-dihydroquinazoline e-7-amide (Compound 52)
N- (3- (4-chlorophenyl) propyl) -6- (3-fluorophenyl) -4-oxo-3, 4-dihydroquinazoline e-7-amide (Compound 53)
6- (3-fluorophenyl) -4-oxo-N- (3-phenylpropyl) -3, 4-dihydroquinazoline e-7-amide (compound 54)
6- (3-chlorophenyl) -4-oxo-N- (3-phenylpropyl) -3, 4-dihydroquinazoline e-7-amide (Compound 55)
6- (3-chlorophenyl) -N- (3- (4-chlorophenyl) propyl) -4-oxo-3, 4-dihydroquinazoline e-7-amide (Compound 56)
4-oxo-N- (3-phenylpropyl) -6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline e-7-amide (compound 57)
N- (3- (4-chlorophenyl) propyl) -4-oxo-6- (3- (trifluoromethyl) phenyl) -3, 4-dihydroquinazoline e-7-amide (compound 58)
6- (3-fluorophenyl) -4-oxo-N- (2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) -3, 4-dihydroquinazoline e-7-amide (compound 59)
Methyl 2- (6- (3-chlorophenyl) -7- ((3- (4-chlorophenyl) propyl) carbamoyl) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 60)
Methyl 2- (6- (3-fluorophenyl) -4-oxo-7- ((2- ((4- (trifluoromethyl) benzyl) oxa) ethyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (Compound 61)
Methyl 2- (6- (3-fluorophenyl) -4-oxo-7- ((3-phenylpropyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (Compound 62)
Methyl 2- (7- ((3- (4-chlorophenyl) propyl) carbamoyl) -6- (3-fluorophenyl) -4-oxoquinazolin-3 (4H) -yl) acetate (Compound 63)
Methyl 2- (6- (3-chlorophenyl) -4-oxo-7- ((3- (4- (trifluoromethyl) phenoxy) propyl) carbamoyl) quinazolin-3 (4H) -yl) acetate (Compound 64)
6- (3-chlorophenyl) -4-oxo-N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (Compound 65)
2- (6- (3-chlorophenyl) -4-oxo-7- ((3- (4- (trifluoromethyl) phenoxy) propyl) carbamoyl) quinazolin-3 (4H) -yl) acetic acid (Compound 66)
6- (3-chlorophenyl) -4-oxo-3- (2-oxo-2- (piperidin-1-yl) ethyl) -N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (Compound 67)
6- (3-chlorophenyl) -3- (2- (3, 3-difluoropiperidin-1-yl) -2-oxoethyl) -4-oxo-N- (3- (4- (trifluoromethyl) phenoxy) propyl) -3, 4-dihydroquinazoline e-7-amide (compound 68)
N- (6- (4- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 69)
N- (6- (3- (dimethylamino) phenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 70)
N- (6- (3-Morpholphenoxy) -4-oxo-3- (2-oxo-2- (prop-2-ynyl-1-amino) ethyl) -3, 4-dihydroquinazolin-7-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (Compound 71).
7. A pharmaceutical composition comprising an effective amount of a compound of any one of claims 1-6 and a pharmaceutically acceptable carrier.
8. Use of a compound according to any one of claims 1 to 6 for the manufacture of a medicament for the prophylaxis or treatment of immunoinflammatory disorders and related conditions thereof.
9. The use according to claim, wherein the immunoinflammatory related disorder is a tumor.
10. The use according to claim 9, wherein said tumor is selected from the group consisting of melanoma, oral epidermoid carcinoma, esophageal carcinoma, gastric carcinoma, lung carcinoma, breast carcinoma, renal carcinoma, liver carcinoma, cervical carcinoma, ovarian carcinoma, pancreatic carcinoma, prostate carcinoma, colon carcinoma, and bladder carcinoma.
CN202010146043.6A 2020-03-05 2020-03-05 Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway Pending CN113354590A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010146043.6A CN113354590A (en) 2020-03-05 2020-03-05 Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010146043.6A CN113354590A (en) 2020-03-05 2020-03-05 Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway

Publications (1)

Publication Number Publication Date
CN113354590A true CN113354590A (en) 2021-09-07

Family

ID=77523545

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010146043.6A Pending CN113354590A (en) 2020-03-05 2020-03-05 Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway

Country Status (1)

Country Link
CN (1) CN113354590A (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101652353A (en) * 2007-04-11 2010-02-17 Cv医药有限公司 3-hydrogen quinazoline-4-one derivatives as stearoyl coa desaturase inhibitors
CN101801937A (en) * 2007-06-22 2010-08-11 艾科尔公司 Quinazolinone compounds and methods of use thereof
CN102050793A (en) * 2009-11-03 2011-05-11 中国医学科学院药物研究所 4(3H) quinazolinone derivatives with antitumor activity
CN106831614A (en) * 2017-01-09 2017-06-13 清华大学 Substituted benzodiazepine * heterocycle compound and its production and use

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101652353A (en) * 2007-04-11 2010-02-17 Cv医药有限公司 3-hydrogen quinazoline-4-one derivatives as stearoyl coa desaturase inhibitors
CN101801937A (en) * 2007-06-22 2010-08-11 艾科尔公司 Quinazolinone compounds and methods of use thereof
CN102050793A (en) * 2009-11-03 2011-05-11 中国医学科学院药物研究所 4(3H) quinazolinone derivatives with antitumor activity
CN106831614A (en) * 2017-01-09 2017-06-13 清华大学 Substituted benzodiazepine * heterocycle compound and its production and use

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
HAO WU ET AL.: "Parallel Solution Phase Synthesis of 3,6,7 - 4(3H)-Quinazolinones and Evaluation of Their Antitumor Activities against Human Cancer", 《J. COMB. CHEM.》 *
JINGSHU YANG ET AL.: "Cp*Co III -catalyzed formal [4+2] cycloaddition of benzamides to afford quinazolinone derivatives", 《CHEM. COMMUN.》 *
XIAOLI ZHOU ET AL.: "Quinazoline-2,4(1 H, 3 H )-diones inhibit the growth of multiple human tumor cell lines", 《MOL DIVERS》 *
YAO MA ET AL.: "Nonpeptidic quinazolinone derivatives as dual nucleotide-binding oligomerization domain-like receptor 1/2 antagonists for adjuvant cancer chemotherapy", 《EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY》 *

Similar Documents

Publication Publication Date Title
TWI299731B (en) Selected fused pyrrolocarbazoles
EP1741714B1 (en) Heterocyclic compound and anti-malignant-tumor agent containing the same as active ingredient
EP1870414A1 (en) Thienopyridine derivative, or quinoline derivative, or quinazoline derivative, having c-met autophosphorylation inhibiting potency
AU2014240003B2 (en) Coumarin derivatives and methods of use in treating hyperproliferative diseases
MX2012009541A (en) Cyclobutane and methylcyclobutane derivatives as janus kinase inhibitors.
WO2013013614A1 (en) 4-(3-heteroarylarylamino)quinazoline and 1-(3-heteroarylarylamino)isoquinoline as hedgehog pathway inhibitor and use thereof
CN109574936B (en) Hydroxamic acid compound with HDAC6 inhibitory activity and application thereof
KR20180066985A (en) Thienopyrimidine derivative and use thereof
WO2015058661A1 (en) Bcr-abl kinase inhibitor and application thereof
CN111989332B (en) Macrocyclic compounds as CDK inhibitors, their preparation and their use in medicine
JP2003533515A (en) Modulator of TNFα signaling
JP2007538102A (en) 5-anilino-4-heteroarylpyrazole derivatives useful for the treatment of diabetes
RU2288918C2 (en) Derivatives of n-triazolylmethylpiperazine, method for their preparing, medicinal agent, derivatives of piperazine
TWI614251B (en) Pyrazolo[4,3-c]quinoline derivatives inhibition β-glucuronidase
CN109574920B (en) 3-nitrile-6-cyclopropyl pyridine IDO1 inhibitor and application thereof
EP3950677A1 (en) Quinolyl-containing compound and pharmaceutical composition, and use thereof
CN113354590A (en) Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway
WO2023130539A1 (en) Triazolopyrimidinol compounds as well as preparation method therefor and use thereof
WO2009097709A1 (en) Pyrazolopyrimidinone-containing phenyl guanidine derivatives, pharmaceutical compositions containing them, process for their preparation and their use
CN109748914B (en) Pyridopyrimidine compound and application thereof
WO2016127949A1 (en) Pyrimidine derivative as inhibitor for t790 mutation
CN111630048B (en) Amidine and guanidine derivatives, preparation method and medical application thereof
JP7110335B2 (en) Pyridoquinazoline derivatives useful as protein kinase inhibitors
CN109384793B (en) Thiol compound with HDAC6 inhibitory activity and application thereof
CN113214230B (en) 2-substituted pyrazol amino-4-substituted amino-5-pyrimidine formamide compound, composition and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
WD01 Invention patent application deemed withdrawn after publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20210907